US11439642B2 - Substituted 5-fluoro-1H-pyrazolopyridines and their use - Google Patents

Substituted 5-fluoro-1H-pyrazolopyridines and their use Download PDF

Info

Publication number
US11439642B2
US11439642B2 US16/919,385 US202016919385A US11439642B2 US 11439642 B2 US11439642 B2 US 11439642B2 US 202016919385 A US202016919385 A US 202016919385A US 11439642 B2 US11439642 B2 US 11439642B2
Authority
US
United States
Prior art keywords
compound
treating
formula
inhibitors
salt
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US16/919,385
Other versions
US20200397785A1 (en
Inventor
Markus Follmann
Johannes-Peter Stasch
Gorden Redlich
Jens Ackerstaff
Nils Griebenow
Walter Kroh
Andreas Knorr
Eva-Maria Becker
Frank Wunder
Volkhart Min-Jian Li
Elke Hartmann
Joachim Mittendorf
Karl-Heinz Schlemmer
Rolf Jautelat
Donald Bierer
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Adverio Pharma GmbH
Original Assignee
Adverio Pharma GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Adverio Pharma GmbH filed Critical Adverio Pharma GmbH
Priority to US16/919,385 priority Critical patent/US11439642B2/en
Publication of US20200397785A1 publication Critical patent/US20200397785A1/en
Assigned to BAYER PHARMA AKTIENGESELLSCHAFT reassignment BAYER PHARMA AKTIENGESELLSCHAFT CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: BAYER SCHERING PHARMA AKTIENGESELLSCHAFT
Assigned to ADVERIO PHARMA GMBH reassignment ADVERIO PHARMA GMBH NUNC PRO TUNC ASSIGNMENT (SEE DOCUMENT FOR DETAILS). Assignors: BAYER INTELLECTUAL PROPERTY GMBH
Assigned to BAYER INTELLECTUAL PROPERTY GMBH reassignment BAYER INTELLECTUAL PROPERTY GMBH NUNC PRO TUNC ASSIGNMENT (SEE DOCUMENT FOR DETAILS). Assignors: BAYER PHARMA AKTIENGESELLSCHAFT
Assigned to BAYER SCHERING PHARMA AKTIENGESELLSCHAFT reassignment BAYER SCHERING PHARMA AKTIENGESELLSCHAFT ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FOLLMANN, MARKUS, LI, VOLKHART MIN-JIAN, GRIEBENOW, NILS, ACKERSTAFF, JENS, BECKER, EVA-MARIA, SCHLEMMER, KARL-HEINZ, WUNDER, FRANK, BIERER, DONALD, HARTMANN, ELKE, JAUTELAT, ROLF, KNORR, ANDREAS, KROH, WALTER, MITTENDORF, JOACHIM, REDLICH, GORDEN, STASCH, JOHANNES-PETER
Priority to US17/820,667 priority patent/US20220409618A1/en
Application granted granted Critical
Publication of US11439642B2 publication Critical patent/US11439642B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C309/00Sulfonic acids; Halides, esters, or anhydrides thereof
    • C07C309/01Sulfonic acids
    • C07C309/02Sulfonic acids having sulfo groups bound to acyclic carbon atoms
    • C07C309/03Sulfonic acids having sulfo groups bound to acyclic carbon atoms of an acyclic saturated carbon skeleton
    • C07C309/04Sulfonic acids having sulfo groups bound to acyclic carbon atoms of an acyclic saturated carbon skeleton containing only one sulfo group
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C309/00Sulfonic acids; Halides, esters, or anhydrides thereof
    • C07C309/01Sulfonic acids
    • C07C309/02Sulfonic acids having sulfo groups bound to acyclic carbon atoms
    • C07C309/03Sulfonic acids having sulfo groups bound to acyclic carbon atoms of an acyclic saturated carbon skeleton
    • C07C309/05Sulfonic acids having sulfo groups bound to acyclic carbon atoms of an acyclic saturated carbon skeleton containing at least two sulfo groups bound to the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C57/00Unsaturated compounds having carboxyl groups bound to acyclic carbon atoms
    • C07C57/02Unsaturated compounds having carboxyl groups bound to acyclic carbon atoms with only carbon-to-carbon double bonds as unsaturation
    • C07C57/13Dicarboxylic acids
    • C07C57/145Maleic acid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the present application relates to novel substituted 5-fluoro-1H-pyrazolopyridines, to processes for their preparation, to their use alone or in combinations for the treatment and/or prophylaxis of diseases, and to their use for producing medicaments for the treatment and/or prophylaxis of diseases, in particular for the treatment and/or prophylaxis of cardiovascular disorders.
  • cyclic guanosine monophosphate cGMP
  • NO nitrogen monoxide
  • GTP guanosine triphosphate
  • the soluble guanylate cyclases consist of two subunits and very probably contain one haem per heterodimer, which is part of the regulatory site. It is of central importance for the activation mechanism. NO is able to bind to the iron atom of haem and thus markedly increase the activity of the enzyme. Haem-free preparations cannot, by contrast, be stimulated by NO. Carbon monoxide (CO) is also able to attach to the central iron atom of haem, but the stimulation by CO is distinctly less than that by NO.
  • CO Carbon monoxide
  • guanylate cyclase plays an important role in various physiological processes, in particular in the relaxation and proliferation of smooth muscle cells, in platelet aggregation and platelet adhesion and in neuronal signal transmission, and also in disorders which are based on a disturbance of the abovementioned processes.
  • the NO/cGMP system may be suppressed, which may lead for example to high blood pressure, platelet activation, increased cellular proliferation, endothelial dysfunction, atherosclerosis, angina pectoris, heart failure, myocardial infarction, thromboses, stroke and sexual dysfunction.
  • WO 00/06569 discloses fused pyrazole derivatives and WO 03/095451 carbamate-substituted 3-pyrimidinylyrazolopyridines as stimulators of soluble guanylate cyclase.
  • the present invention provides compounds of the general formula (I)
  • Compounds according to the invention are the compounds of the formula (I) and the N-oxides, salts, solvates and solvates of the N-oxides and salts thereof, the compounds, encompassed by formula (I), of the formulae specified hereinafter and the N-oxides, salts, solvates and solvates of the N-oxides and salts thereof, and the compounds encompassed by formula (I) and specified hereinafter as working examples and the N-oxides, salts, solvates and solvates of the N-oxides and salts thereof, to the extent that the compounds encompassed by formula (I) and specified hereinafter are not already N-oxides, salts, solvates and solvates of the N-oxides and salts.
  • preferred salts are physiologically acceptable salts of the compounds according to the invention. Also encompassed are salts which are not themselves suitable for pharmaceutical applications but can be used, for example, for isolation or purification of the compounds according to the invention.
  • Physiologically acceptable salts of the compounds according to the invention include acid addition salts of mineral acids, carboxylic acids and sulphonic acids, for example salts of hydrochloric acid, hydrobromic acid, sulphuric acid, phosphoric acid, methanesulphonic acid, ethanesulphonic acid, toluenesulphonic acid, benzenesulphonic acid, naphthalenedisulphonic acid, formic acid, acetic acid, trifluoroacetic acid, propionic acid, lactic acid, tartaric acid, malic acid, citric acid, fumaric acid, maleic acid and benzoic acid.
  • Physiologically acceptable salts of the compounds according to the invention also include salts of conventional bases, by way of example and with preference alkali metal salts (e.g. sodium and potassium salts), alkaline earth metal salts (e.g. calcium and magnesium salts) and ammonium salts derived from ammonia or organic amines having 1 to 16 carbon atoms, by way of example and with preference ethylamine, diethylamine, triethylamine, ethyldiisopropylamine, monoethanolamine, diethanolamine, triethanolamine, dicyclohexylamine, dimethylaminoethanol, procaine, dibenzylamine, N-methylmorpholine, arginine, lysine, ethylenediamine and N-methylpiperidine.
  • alkali metal salts e.g. sodium and potassium salts
  • alkaline earth metal salts e.g. calcium and magnesium salts
  • ammonium salts derived from ammonia or organic amines
  • solvates refer to those forms of the compounds according to the invention which, in the solid or liquid state, form a complex by coordination with solvent molecules. Hydrates are a specific form of the solvates in which the coordination is with water. Preferred solvates in the context of the present invention are hydrates.
  • the compounds according to the invention may exist in different stereoisomeric forms, i.e. in the form of configurational isomers or if appropriate also as conformational isomers (enantiomers and/or diastereomers, including those in the case of atropisomers).
  • the present invention therefore encompasses the enantiomers or diastereomers and the respective mixtures thereof.
  • the stereoisomerically uniform constituents can be isolated from such mixtures of enantiomers and/or diastereomers in a known manner; chromatography processes are preferably used for this, in particular HPLC chromatography on an achiral or chiral phase.
  • the present invention also encompasses all suitable isotopic variants of the compounds according to the invention.
  • An isotopic variant of a compound according to the invention is understood here to mean a compound in which at least one atom within the compound according to the invention compound has been exchanged for another atom of the same atomic number, but with a different atomic mass than the atomic mass which usually or predominantly occurs in nature.
  • isotopes which can be incorporated into a compound according to the invention are those of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulphur, fluorine, chlorine, bromine and iodine, such as 2 H (deuterium), 3 H (tritium), 13 c, 14 C, 15 N, 17 O, 18 O, 32 P, 33 P, 33 S, 34 S, 35 S, 36 S, 18 F, 36 Cl, 82 Br, 123 I, 124 I, 129 I and 131 I.
  • Particular isotopic variants of a compound according to the invention may be beneficial, for example, for the examination of the mechanism of action or of the active compound distribution in the body; due to comparatively easy preparability and detectability, especially compounds labelled with 3 H or 14 C isotopes are suitable for this purpose.
  • the incorporation of isotopes, for example of deuterium can lead to particular therapeutic benefits as a consequence of greater metabolic stability of the compound, for example an extension of the half-life in the body or a reduction in the active dose required; such modifications of the compounds according to the invention may therefore in some cases also constitute a preferred embodiment of the present invention.
  • Isotopic variants of the compounds according to the invention can be prepared by processes known to those skilled in the art, for example by the methods described below and the methods described in the working examples, by using corresponding isotopic modifications of the particular reagents and/or starting compounds therein.
  • the present invention also encompasses prodrugs of the compounds according to the invention.
  • prodrugs refers to compounds which for their part can be biologically active or inactive, but are converted (for example metabolically or hydrolytically) into compounds according to the invention during their dwell time in the body.
  • alkyl represents a straight-chain or branched alkyl radical having 1 to 4 carbon atoms.
  • the following may be mentioned by way of example and by way of preference: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, 1-methylpropyl, tert-butyl.
  • halogen represents fluorine, chlorine, bromine and iodine.
  • radicals in the compounds according to the invention are substituted, the radicals may be mono- or polysubstituted, unless specified otherwise.
  • all radicals which occur more than once are defined independently of one another. Substitution by one, two or three identical or different substituents is preferred.
  • methyl may be substituted by a trifluoromethyl substituent, and their salts, solvates and solvates of the salts.
  • the invention further provides a process for preparing the compounds of the formula (I) according to the invention, characterized in that the compound of the formula (II)
  • Inert solvents for process step (II)+(III) ⁇ (IV) are, for example, alcohols such as methanol, ethanol, n-propanol, isopropanol, n-butanol or tert-butanol, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, hydrocarbons such as benzene, xylene, toluene, hexane, cyclohexane or mineral oil fractions, or other solvents such as dimethylformamide (DMF), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU), dimethylacetamide, N-methylpyrrolidone (NMP), pyridine, acetonitrile, sulpholane or else water. It is equally possible to use mixtures of the solvents mentioned. Preference is given to dioxan
  • Suitable palladium catalysts for process step (II)+(III) ⁇ (IV) are, for example, palladium on activated carbon, palladium(II) acetate, tetrakis(triphenylphosphine)palladium(0), bis(triphenylphosphine)palladium(II) chloride, bis(acetonitrile)palladium(II) chloride and [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II)/dichloromethane complex, if appropriate in combination with additional phosphane ligands such as, for example, (2-biphenyl)di-tert-butylphosphine, dicyclohexyl[2′,4′,6′-tris(1-methylethyl)biphenyl-2-yl]phosphane (XPHOS), bis(2-phenylphosphinophenyl) ether
  • the reaction (II)+(III) ⁇ (IV) is generally carried out in a temperature range of from +20° C. to +180° C., prepferably from +50° C. to +120° C., if appropriate in a microwave.
  • the reaction can be carried out at atmospheric, elevated or reduced pressure (for example from 0.5 to 5 bar).
  • the reaction is generally carried out at atmospheric pressure.
  • the reductions (IV) ⁇ (V) and (IX) ⁇ (V) are carried out in the presence of a suitable catalyst in an inert solvent in a temperature range of from +20° C. to +40° C. under hydrogen of atmospheric pressure.
  • Inert solvents for the reductions (IV) ⁇ (V) and (IX) ⁇ (V) are, for example, alcohols such as methanol, ethanol, n-propanol, isopropanol, n-butanol or tert-butanol, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, or other solvents such as dimethylformamide (DMF), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU), N-methylpyrrolidone (NMP), pyridine, acetonitrile or else water. It is equally possible to use mixtures of the solvents mentioned. Preference is given to DMF and pyridine.
  • Suitable catalysts for the reactions (IV) ⁇ (V) and (IX) ⁇ (V) are, for example, palladium on activated carbon, platinum on carbon, palladium hydroxide or Raney nickel.
  • the reductions (IV) ⁇ (V) and (IX) ⁇ (V) can be carried out using a metal or metal salt such as, for example, iron, zinc or tin(II) chloride in a suitable acid such as, for example, hydrogen chloride/hydrochloric acid, sulphuric acid, phosphoric acid or acetic acid in a temperature range of from +20° C. to +140° C.
  • a metal or metal salt such as, for example, iron, zinc or tin(II) chloride in a suitable acid such as, for example, hydrogen chloride/hydrochloric acid, sulphuric acid, phosphoric acid or acetic acid in a temperature range of from +20° C. to +140° C.
  • Inert solvents for process step (V) ⁇ (I-A) are, for example, alcohols such as methanol, ethanol, n-propanol, isopropanol, n-butanol or tert-butanol, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, trichloroethylene or chlorobenzene, hydrocarbons such as benzene, xylene, toluene, hexane, cyclohexane or mineral oil fractions, or other solvents such as dimethylformamide (DMF), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU), N-methylpyrrolidone (NMP), acetonitrile or
  • Suitable bases for the process step (V) ⁇ (I-A) are alkali metal hydrides such as sodium hydride, alkali metal hydroxides such as, for example, lithium hydroxide, sodium hydroxide or potassium hydroxide, alkali metal carbonates such as lithium carbonate, sodium carbonate, potassium carbonate or caesium carbonate, alkali metal bicarbonates such as sodium bicarbonate or potassium bicarbonate, alkali metal alkoxides such as sodium methoxide or potassium methoxide, sodium ethoxide or potassium ethoxide or potassium tert-butoxide, or organic amines such as triethylamine, diisopropylethylamine, pyridine, 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU) or 1,5-diazabicyclo[4.3.0]non-5-ene (DBN). Preference is given to pyridine.
  • alkali metal hydrides such as sodium hydride
  • the reaction (V) ⁇ (I-A) is generally carried out in a temperature range of from ⁇ 10° C. to +30° C., preferably from 0° C. to +20° C.
  • the reaction can be carried out at atmospheric, elevated or reduced pressure (for example from 0.5 to 5 bar).
  • the reaction is generally carried out at atmospheric pressure.
  • Inert solvents for process step (II) ⁇ (VI) are, for example, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, hydrocarbons such as benzene, xylene, toluene, hexane, cyclohexane or mineral oil fractions, or other solvents such as dimethylformamide (DMF), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU), N-methylpyrrolidone (NMP), pyridine or acetonitrile. It is equally possible to use mixtures of the solvents mentioned. Preference is given to DMSO.
  • the reaction (II) ⁇ (VI) is generally carried out in a temperature range of from +20° C. to +180° C., preferably from +100° C. to +160° C., if appropriate in a microwave.
  • the reaction can be carried out at atmospheric, elevated or reduced pressure (for example from 0.5 to 5 bar).
  • the reaction is generally carried out at atmospheric pressure.
  • reaction (VI) ⁇ (VII) is carried out using methods known to the person skilled in the art in a two-step process initially with formation of the imino ester using sodium methoxide in methanol at from 0° C. to +40° C. and subsequent nucleophilic addition of an ammonia equivalent such as, for example, ammonia or ammonium chloride in acetic acid with formation of the amidine (VII) at from +50 to +150° C.
  • an ammonia equivalent such as, for example, ammonia or ammonium chloride in acetic acid
  • Inert solvents for process step (VII)+(VIII) ⁇ (IX) are alcohols such as methanol, ethanol, n-propanol, isopropanol, n-butanol or tert-butanol, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, hydrocarbons such as benzene, xylene, toluene, hexane, cyclohexane or mineral oil fractions, or other solvents such as dimethylformamide (DMF), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU), N-methylpyrrolidone (NMP), pyridine, acetonitrile or else water. It is equally possible to use mixtures of the solvents mentioned. Preference is given to DMF.
  • Suitable bases for the process step (VII)+(VIII) ⁇ (IX) are alkali metal hydroxides such as, for example, lithium hydroxide, sodium hydroxide or potassium hydroxide, alkali metal carbonates such as lithium carbonate, sodium carbonate, potassium carbonate or caesium carbonate, alkali metal bicarbonates such as sodium bicarbonate or potassium bicarbonate, alkali metal alkoxides such as sodium methoxide or potassium methoxide, sodium ethoxide or potassium ethoxide or potassium tert-butoxide, or organic amines such as triethylamine, diisopropylethylamine, pyridine, 1,8-diazabicyclo [5.4.0]undec-7-ene (DBU) or 1,5-diazabicyclo[4.3.0]non-5-ene (DBN). Preference is given to triethylamine.
  • alkali metal hydroxides such as, for example, lithium hydroxide, sodium hydrox
  • the reaction (VII)+(VIII) ⁇ (IX) is generally carried out in a temperature range of from +20° C. to +150° C., preferably from +80° C. to +120° C., if appropriate in a microwave.
  • the reaction can be carried out at atmospheric, elevated or reduced pressure (for example from 0.5 to 5 bar).
  • the reaction is generally carried out at atmospheric pressure.
  • the compound of the formula (VIII) can be prepared analogously to the literature L. F. Cavalieri, J. F. Tanker, A. Bendich, J. Am. Chem. Soc., 1949, 71, 533.
  • Inert solvents for the reaction (I-A) ⁇ (I-B) are, for example, halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, trichloroethylene or chlorobenzene, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, or other solvents such as dimethylformamide (DMF), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU), N-methylpyrrolidone (NMP), pyridine or acetonitrile. Preference is given to tetrahydrofuran.
  • halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, trichloroethylene or chlorobenzene
  • ethers such as diethyl ether, diox
  • Suitable bases for the process step (I-A) ⁇ (I-B) are alkali metal hydrides such as potassium hydride or sodium hydride, alkali metal carbonates such as lithium carbonate, sodium carbonate, potassium carbonate or caesium carbonate, alkali metal bicarbonates such as sodium bicarbonate or potassium bicarbonate, alkali metal alkoxides such as sodium methoxide or potassium methoxide, sodium ethoxide or potassium ethoxide or potassium tert-butoxide, amides such as sodium amide, lithium bis(trimethylsilyl)amide, sodium bis(trimethylsilyl)amide or potassium bis(trimethylsilyl)amide or lithium diisopropylamide, organometallic compounds such as butyllithium or phenyllithium, or organic amines such as triethylamine, diisopropylethylamine, pyridine, 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU)
  • the reaction (I-A) ⁇ (I-B) is generally carried out in a temperature range of from ⁇ 78° C. to +40° C., preferably from 0° C. to +20° C.
  • the reaction can be carried out at atmospheric, elevated or reduced pressure (for example from 0.5 to 5 bar).
  • the reaction is generally carried out at atmospheric pressure.
  • the compound of the formula (II) can be prepared by cyclizing the compound of the formula (X)
  • Inert solvents for process step (X) ⁇ (XI) are alcohols such as methanol, ethanol, n-propanol, isopropanol, n-butanol, tert-butanol or 1,2-ethanediol, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, hydrocarbons such as benzene, xylene, toluene, hexane, cyclohexane or mineral oil fractions, or other solvents such as dimethylformamide (DMF), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU), N-methylpyrrolidone (NMP), pyridine, acetonitrile or else water. It is equally possible to use mixtures of the solvents mentioned. 1,2-Ethanediol is preferred.
  • the reaction (X) ⁇ (XI) is generally carried out in a temperature range of from +60° C. to +200° C., preferably from +120° C. to +180° C.
  • the reaction can be carried out at atmospheric, elevated or reduced pressure (for example from 0.5 to 5 bar).
  • the reaction is generally carried out at atmospheric pressure.
  • Inert solvents for the reaction (XI) ⁇ (XII) are, for example, halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, trichloroethylene or chlorobenzene, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, or other solvents such as dimethylformamide (DMF), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU), N-methylpyrrolidone (NMP), pyridine or acetonitrile. Preference is given to DMF.
  • halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, trichloroethylene or chlorobenzene
  • ethers such as diethyl ether, dioxane, tetra
  • Suitable Lewis acids for the process step (XI) ⁇ (XII) are boron trifluoride/diethyl ether complex, cerium(IV) ammonium nitrate (CAN), tin(II) chloride, lithium perchlorate, zinc(II) chloride, indium(III) chloride or indium(III) bromide. Preference is given to boron trifluoride/diethyl ether complex.
  • the reaction (XI) ⁇ (XII) is generally carried out in a temperature range of from ⁇ 78° C. to +40° C., preferably from 0° C. to +20° C.
  • the reaction can be carried out at atmospheric, elevated or reduced pressure (for example from 0.5 to 5 bar).
  • the reaction is generally carried out at atmospheric pressure.
  • Inert solvents for the reaction (XII)+(XIII) ⁇ (II) are, for example, halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, trichloroethylene or chlorobenzene, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, or other solvents such as dimethylformamide (DMF), dimethyl sulphoxide (DMSO), N,N′-dimcthylpropylcncurca (DMPU), N-methylpyrrolidone (NMP), pyridine or acetonitrile. Preference is given to DMF.
  • halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, trichloroethylene or chlorobenzene
  • ethers such as diethyl ether,
  • Suitable bases for the process step (XII)+(XIII) ⁇ (II) are alkali metal hydrides such as potassium hydride or sodium hydride, alkali metal carbonates such as lithium carbonate, sodium carbonate, potassium carbonate or caesium carbonate, alkali metal bicarbonates such as sodium bicarbonate or potassium bicarbonate, alkali metal alkoxides such as sodium methoxide or potassium methoxide, sodium ethoxide or potassium ethoxide or potassium tert-butoxide, amides such as sodium amide, lithium bis(trimethylsilyl)amide, sodium bis(trimethylsilyl)amide or potassium bis(trimethylsilyl)amide or lithium diisopropylamide, organometallic compounds such as butyllithium or phenyllithium, or organic amines such as triethylamine, diisopropylethylamine, pyridine, 1,8-diazabicyclo[5.4.0]undec-7-ene
  • the reaction (XII)+(XIII) ⁇ (II) is generally carried out in a temperature range of from 0° C. to +60° C., preferably from +10° C. to +25° C.
  • the reaction can be carried out at atmospheric, elevated or reduced pressure (for example from 0.5 to 5 bar).
  • the reaction is generally carried out at atmospheric pressure.
  • the present invention furthermore provides the compound:
  • the present invention furthermore provides the compound:
  • the compound of the formula (X) is known from the literature [cf., for example, Winn M., J. Med. Chem. 1993, 36, 2676-7688; EP 634 413-A1; CN 1613849-A; EP 1626045-A1; WO 2009/018415] and can be prepared in analogy to literature processes or as shown in the synthesis scheme below (Scheme 5):
  • the compounds according to the invention act as stimulators of soluble guanylate cyclase and have an identical or improved therapeutic profile compared to compounds known from the prior art, such as, for example, with respect to their in vivo properties such as, for example, their pharmacokinetic and pharmacodynamic behaviour and/or their dose-activity relationship and/or their safety profile. They are therefore suitable for the treatment and/or prophylaxis of diseases in man and animals.
  • the compounds according to the invention lead to vasorelaxation, to an inhibition of platelet aggregation and to a reduction in blood pressure, and also to an increase in coronary blood flow. These effects are mediated via direct stimulation of soluble guanylate cyclase and intracellular cGMP increase. Moreover, the compounds according to the invention enhance the effect of substances increasing the cGMP concentration, such as, for example, EDRF (endothelium-derived relaxing factor), NO donors, protoporphyrin IX, arachidonic acid or phenylhydrazine derivatives.
  • EDRF endothelium-derived relaxing factor
  • the compounds according to the invention are suitable for the treatment and/or prophylaxis of cardiovascular, pulmonary, thromboembolic and fibrotic disorders.
  • the compounds according to the invention can be used in medicaments for the treatment and/or prophylaxis of cardiovascular disorders such as, for example, hypertension, acute and chronic heart failure, coronary heart disease, stable and unstable angina pectoris, peripheral and cardiac vascular disorders, arrhythmias, atrial and ventricular arrhythmias and impaired conduction such as, for example, atrioventricular blocks degrees I-III (AB block supraventricular tachyarrhythmia, atrial fibrillation, atrial flutter, ventricular fibrillation, ventricular flutter, ventricular tachyarrhythmia, Torsade de pointes tachycardia, atrial and ventricular extrasystoles, AV-junctional extrasystoles, Sick-Sinus syndrome, syncopes, AV-nodal re-entry tachycardia, Wolff-Parkinson-White syndrome, of acute coronary syndrome (ACS), autoimmune cardiac disorders (pericarditis, endocarditis, end
  • heart failure also includes more specific or related types of disease, such as acute decompensated heart failure, right heart failure, left heart failure, global failure, ischaemic cardiomyopathy, dilated cardiomyopathy, hypertrophic cardiomyopathy, idiopathic cardiomyopathy, congenital heart defects, heart valve defects, heart failure associated with heart valve defects, mitral stenosis, mitral insufficiency, aortic stenosis, aortic insufficiency, tricuspid stenosis, tricuspid insufficiency, pulmonary valve stenosis, pulmonary valve insufficiency, combined heart valve defects, myocardial inflammation (myocarditis), chronic myocarditis, acute myocarditis, viral myocarditis, diabetic heart failure, alcoholic cardiomyopathy, cardiac storage disorders, and diastolic and systolic heart failure.
  • myocardial inflammation myocarditis
  • chronic myocarditis chronic myocarditis
  • the compounds according to the invention can also be used for the treatment and/or prophylaxis of arteriosclerosis, impaired lipid metabolism, hypolipoproteinaemias, dyslipidaemias, hypertriglyceridaemias, hyperlipidaemias, hypercholesterolaemias, abetalipoproteinaemias, sitosterolaemia, xanthomatosis, Tangier disease, adipositas, obesity and of combined hyperlipidaemias and metabolic syndrome.
  • the compounds according to the invention can additionally be used for the treatment and/or prophylaxis of primary and secondary Raynaud's phenomenon, of microcirculation impairments, claudication, peripheral and autonomic neuropathies, diabetic microangiopathies, diabetic retinopathy, diabetic ulcers on the extremities, gangrene, CREST syndrome, erythematosis, onychomycosis, rheumatic disorders and for promoting wound healing.
  • the compounds according to the invention are furthermore suitable for treating urological disorders such as, for example, benign prostate syndrome (BPS), benign prostate hyperplasia (BPH), benign prostate enlargement (BPE), bladder outlet obstruction (BOO), lower urinary tract syndromes (LUTS, including Feline Urological Syndrome (FUS)), disorders of the urogenital system including neurogenic over-active bladder (OAB) and (IC), incontinence (UI) such as, for example, mixed urinary incontinence, urge urinary incontinence, stress urinary incontinence or overflow urinary incontinence (MUI, UUI, SUI, OUI), pelvic pain, benign and malignant disorders of the organs of the male and female urogenital system.
  • BPS benign prostate syndrome
  • BPH benign prostate hyperplasia
  • BPE benign prostate enlargement
  • BOO bladder outlet obstruction
  • LUTS lower urinary tract syndromes
  • LUTS lower urinary tract syndromes
  • FUS Feline Urological Syndrome
  • UI incontinence
  • kidney disorders in particular of acute and chronic renal insufficiency and acute and chronic renal failure.
  • renal insufficiency comprises both acute and chronic manifestations thereof, as well as underlying or related kidney diseases such as renal hypoperfusion, intradialytic hypotension, obstructive uropathy, glomerulopathies, glomerulonephritis, acute glomerulonephritis, glomerulosclerosis, tubulointerstitial diseases, nephropathic diseases such as primary and congenital kidney disease, nephritis, immunological kidney diseases such as kidney graft rejection and immunocomplex-induced kidney diseases, nephropathy induced by toxic substances, nephropathy induced by contrast agents, diabetic and non-diabetic nephropathy, pyelonephritis, renal cysts, nephrosclerosis, hypertensive nephrosclerosis
  • the present invention also comprises the use of the compounds according to the invention for the treatment and/or prophylaxis of sequelae of renal insufficiency, for example pulmonary oedema, heart failure, uraemia, anaemia, electrolyte disturbances (for example hypercalaemia, hyponatraemia) and disturbances in bone and carbohydrate metabolism.
  • sequelae of renal insufficiency for example pulmonary oedema, heart failure, uraemia, anaemia, electrolyte disturbances (for example hypercalaemia, hyponatraemia) and disturbances in bone and carbohydrate metabolism.
  • the compounds according to the invention are also suitable for the treatment and/or prophylaxis of asthmatic disorders, pulmonary arterial hypertension (PAH) and other forms of pulmonary hypertension (PH) including left-heart disease, HIV, sickle cell anaemia, thromboembolisms (CTEPH), sarkoidosis, COPD or pulmonary fibrosis-associated pulmonary hypertension, chronic-obstructive pulmonary disease (COPD), acute respiratory distress syndrome (ARDS), acute lung injury (ALI), alpha-1-antitrypsin deficiency (AATD), pulmonary fibrosis, pulmonary emphysema (for example pulmonary emphysema induced by cigarette smoke) and cystic fibrosis (CF).
  • PAH pulmonary arterial hypertension
  • PH pulmonary hypertension
  • COPD chronic-obstructive pulmonary disease
  • ARDS acute respiratory distress syndrome
  • ALI acute lung injury
  • AATD alpha-1-antitrypsin deficiency
  • the compounds described in the present invention also represent active compounds for controlling central nervous system diseases characterized by disturbances of the NO/cGMP system. They are suitable in particular for improving perception, concentration, learning or memory after cognitive impairments like those occurring in particular in association with situations/diseases/syndromes such as mild cognitive impairment, age-associated learning and memory impairments, age-associated memory losses, vascular dementia, craniocerebral trauma, stroke, dementia occurring after strokes (post stroke dementia), post-traumatic craniocerebral trauma, general concentration impairments, concentration impairments in children with learning and memory problems, Alzheimer's disease, Lewy body dementia, dementia with degeneration of the frontal lobes including Pick's syndrome, Parkinson's disease, progressive nuclear palsy, dementia with corticobasal degeneration, amyolateral sclerosis (ALS), Huntington's disease, demyelination, multiple sclerosis, thalamic degeneration, Creutzfeld-Jacob dementia, HIV dementia, schizophrenia with dementia or Korsakoff's psychosis.
  • the compounds according to the invention are furthermore also suitable for controlling cerebral blood flow and thus represent effective agents for controlling migraine. They are also suitable for the prophylaxis and control of sequelae of cerebral infarct (Apoplexia cerebri) such as stroke, cerebral ischaemias and skull-brain trauma.
  • the compounds according to the invention can likewise be employed for controlling states of pain and tinnitus.
  • the compounds according to the invention have antiinflammatory action and can therefore be used as antiinflammatory agents for the treatment and/or prophylaxis of sepsis (SIRS), multiple organ failure (MODS, MOF), inflammatory disorders of the kidney, chronic intestinal inflammations (IBD, Crohn's disease, UC), pancreatitis, peritonitis, rheumatoid disorders, inflammatory skin diseases and inflammatory eye diseases.
  • SIRS sepsis
  • MODS multiple organ failure
  • IBD chronic intestinal inflammations
  • Crohn's disease UC
  • pancreatitis peritonitis
  • rheumatoid disorders inflammatory skin diseases and inflammatory eye diseases.
  • the compounds according to the invention can also be used for the treatment and/or prophylaxis of autoimmune diseases.
  • the compounds according to the invention are furthermore suitable for the treatment and/or prophylaxis of fibrotic disorders of the internal organs such as, for example, the lung, the heart, the kidney, the bone marrow and in particular the liver, and also dermatological fibroses and fibrotic eye disorders.
  • fibrotic disorders includes in particular the following terms: hepatic fibrosis, cirrhosis of the liver, pulmonary fibrosis, endomyocardial fibrosis, nephropathy, glomerulonephritis, interstitial renal fibrosis, fibrotic damage resulting from diabetes, bone marrow fibrosis and similar fibrotic disorders, scleroderma, morphea, keloids, hypertrophic scarring (also following surgical procedures), naevi, diabetic retinopathy, proliferative vitreoretinopathy and disorders of the connective tissue (for example sarcoidosis).
  • the compounds according to the invention are furthermore suitable for controlling postoperative scarring, for example as a result of glaucoma operations.
  • the compounds according to the invention can also be used cosmetically for ageing and keratinized skin.
  • the compounds according to the invention are suitable for the treatment and/or prophylaxis of hepatitis, neoplasms, osteoporosis, glaucoma and gastroparesis.
  • the present invention further provides for the use of the compounds according to the invention for the treatment and/or prophylaxis of disorders, in particular the disorders mentioned above.
  • the present invention further provides the use of the compounds according to the invention for the treatment and/or prophylaxis of heart failure, angina pectoris, hypertension, pulmonary hypertension, ischaemias, vascular disorders, kidney failure, thromboembolic disorders, fibrotic disorders and arteriosclerosis.
  • the present invention further provides the compounds according to the invention for use in a method for the treatment and/or prophylaxis of heart failure, angina pectoris, hypertension, pulmonary hypertension, ischaemias, vascular disorders, kidney failure, thromboembolic disorders, fibrotic disorders and arteriosclerosis.
  • the present invention further provides for the use of the compounds according to the invention for producing a medicament for treatment and/or prophylaxis of disorders, in particular the disorders mentioned above.
  • the present invention further provides for the use of the compounds according to the invention for producing a medicament for treatment and/or prophylaxis of heart failure, angina pectoris, hypertension, pulmonary hypertension, ischaemias, vascular disorders, kidney failure, thromboembolic disorders, fibrotic disorders and arteriosclerosis.
  • the present invention further provides a method for treatment and/or prophylaxis of disorders, in particular the disorders mentioned above, using an effective amount of at least one of the compounds according to the invention.
  • the present invention further provides a method for treatment and/or prophylaxis of heart failure, angina pectoris, hypertension, pulmonary hypertension, ischaemias, vascular disorders, kidney failure, thromboembolic disorders, fibrotic disorders and arteriosclerosis using an effective amount of at least one of the compounds according to the invention.
  • the compounds according to the invention can be employed alone or, if required, in combination with other active compounds.
  • the present invention further provides medicaments comprising at least one of the compounds according to the invention and one or more further active compounds, especially for the treatment and/or prophylaxis of the aforementioned disorders.
  • suitable active compound combinations include:
  • Agents having antithrombotic activity preferably mean compounds from the group of platelet aggregation inhibitors, of anticoagulants or of profibrinolytic substances.
  • the compounds according to the invention are administered in combination with a platelet aggregation inhibitor such as, by way of example and preferably, aspirin, clopidogrel, ticlopidin or dipyridamol.
  • a platelet aggregation inhibitor such as, by way of example and preferably, aspirin, clopidogrel, ticlopidin or dipyridamol.
  • the compounds according to the invention are administered in combination with a thrombin inhibitor such as, by way of example and preferably, ximelagatran, dabigatran, melagatran, bivalirudin or clexane.
  • a thrombin inhibitor such as, by way of example and preferably, ximelagatran, dabigatran, melagatran, bivalirudin or clexane.
  • the compounds according to the invention are administered in combination with a GPIIb/IIIa antagonist such as, by way of example and preferably, tirofiban or abciximab.
  • a GPIIb/IIIa antagonist such as, by way of example and preferably, tirofiban or abciximab.
  • the compounds according to the invention are administered in combination with a factor Xa inhibitor such as, by way of example and preferably, rivaroxab an (BAY 59-7939), DU-176b, apixab an, otamixab an, fidexab an, razaxab an, fondaparinux, idraparinux, PMD-3112, YM-150, KFA-1982, EMD-503982, MCM-17, MLN-1021, DX 9065a, DPC 906, JTV 803, SSR-126512 or SSR-128428.
  • a factor Xa inhibitor such as, by way of example and preferably, rivaroxab an (BAY 59-7939), DU-176b, apixab an, otamixab an, fidexab an, razaxab an, fondaparinux, idraparinux, PMD-3112, YM-150, KFA
  • the compounds according to the invention are administered in combination with heparin or a low molecular weight (LMW) heparin derivative.
  • LMW low molecular weight
  • the compounds according to the invention are administered in combination with a vitamin K antagonist such as, by way of example and preferably, coumarin.
  • Agents which lower blood pressure are preferably understood to mean compounds from the group of calcium antagonists, angiotensin AII antagonists, ACE inhibitors, endothelin antagonists, renin inhibitors, alpha-receptor blockers, beta-receptor blockers, mineralocorticoid receptor antagonists, and the diuretics.
  • the compounds according to the invention are administered in combination with a calcium antagonist such as, by way of example and preferably, nifedipine, amlodipine, verapamil or diltiazem.
  • a calcium antagonist such as, by way of example and preferably, nifedipine, amlodipine, verapamil or diltiazem.
  • the compounds according to the invention are administered in combination with an alpha-1 receptor blocker such as, by way of example and preferably, prazosin.
  • the compounds according to the invention are administered in combination with a beta receptor blocker such as, by way of example and preferably, propranolol, atenolol, timolol, pindolol, alprenolol, oxprenolol, penbutolol, bupranolol, metipranolol, nadolol, mepindolol, carazalol, sotalol, metoprolol, betaxolol, celiprolol, bisoprolol, carteolol, esmolol, labetalol, carvedilol, adaprolol, landiolol, nebivolol, epanolol or bucindolol.
  • a beta receptor blocker such as, by way of example and preferably, propranolol, atenolol, timolol, pind
  • the compounds according to the invention are administered in combination with an angiotensin AII antagonist such as, by way of example and preferably, losartan, candesartan, valsartan, telmisartan or embusartan.
  • angiotensin AII antagonist such as, by way of example and preferably, losartan, candesartan, valsartan, telmisartan or embusartan.
  • the compounds according to the invention are administered in combination with an ACE inhibitor such as, by way of example and preferably, enalapril, captopril, lisinopril, ramipril, delapril, fosinopril, quinopril, perindopril or trandopril.
  • an ACE inhibitor such as, by way of example and preferably, enalapril, captopril, lisinopril, ramipril, delapril, fosinopril, quinopril, perindopril or trandopril.
  • the compounds according to the invention are administered in combination with an endothelin antagonist such as, by way of example and preferably, bosentan, darusentan, ambrisentan or sitaxsentan.
  • an endothelin antagonist such as, by way of example and preferably, bosentan, darusentan, ambrisentan or sitaxsentan.
  • the compounds according to the invention are administered in combination with a renin inhibitor such as, for example and preferably, aliskiren, SPP-600 or SPP-800.
  • a renin inhibitor such as, for example and preferably, aliskiren, SPP-600 or SPP-800.
  • the compounds according to the invention are administered in combination with a mineralocorticoid receptor antagonist such as, for example and preferably, spironolactone or eplerenone.
  • a mineralocorticoid receptor antagonist such as, for example and preferably, spironolactone or eplerenone.
  • the compounds according to the invention are administered in combination with a loop diuretic such as, for example, furosemide, torasemide, bumetanide and piretanide, with potassium-sparing diuretics such as, for example, amiloride and triamterene, with aldosterone antagonists such as, for example, spironolactone, potassium canrenoate and eplerenone and also thiazide diuretics such as, for example, hydrochlorothiazide, chlorthalidone, xipamide and indapamide.
  • a loop diuretic such as, for example, furosemide, torasemide, bumetanide and piretanide
  • potassium-sparing diuretics such as, for example, amiloride and triamterene
  • aldosterone antagonists such as, for example, spironolactone, potassium canrenoate and eplerenone
  • thiazide diuretics such as, for
  • Active compounds which alter lipid metabolism are preferably understood to mean compounds from the group of CETP inhibitors, thyroid receptor agonists, cholesterol synthesis inhibitors such as HMG-CoA reductase inhibitors or squalene synthesis inhibitors, of ACAT inhibitors, MTP inhibitors, PPAR-alpha, PPAR-gamma and/or PPAR-delta agonists, cholesterol absorption inhibitors, polymeric bile acid adsorbents, bile acid reabsorption inhibitors, lipase inhibitors and lipoprotein(a) antagonists;
  • the compounds according to the invention are administered in combination with a CETP inhibitor such as, by way of example and preferably, dalcetrapib, BAY 60-5521, anacetrapib or CETP vaccine (CETi-1).
  • a CETP inhibitor such as, by way of example and preferably, dalcetrapib, BAY 60-5521, anacetrapib or CETP vaccine (CETi-1).
  • the compounds according to the invention are administered in combination with a thyroid receptor agonist such as, by way of example and preferably, D-thyroxin, 3,5,3′-triiodothyronin (T3), CGS 23425 or axitirome (CGS 26214).
  • a thyroid receptor agonist such as, by way of example and preferably, D-thyroxin, 3,5,3′-triiodothyronin (T3), CGS 23425 or axitirome (CGS 26214).
  • the compounds according to the invention are administered in combination with a HMG-CoA reductase inhibitor from the class of the statins such as, by way of example and preferably, lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, rosuvastatin or pitavastatin.
  • a HMG-CoA reductase inhibitor from the class of the statins such as, by way of example and preferably, lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, rosuvastatin or pitavastatin.
  • the compounds according to the invention are administered in combination with a squalene synthesis inhibitor such as, by way of example and preferably, BMS-188494 or TAK-475.
  • a squalene synthesis inhibitor such as, by way of example and preferably, BMS-188494 or TAK-475.
  • the compounds according to the invention are administered in combination with an ACAT inhibitor such as, by way of example and preferably, avasimibe, melinamide, pactimibe, eflucimibe or SMP-797.
  • an ACAT inhibitor such as, by way of example and preferably, avasimibe, melinamide, pactimibe, eflucimibe or SMP-797.
  • the compounds according to the invention are administered in combination with an MTP inhibitor such as, by way of example and preferably, implitapide, BMS-201038, R-103757 or JTT-130.
  • an MTP inhibitor such as, by way of example and preferably, implitapide, BMS-201038, R-103757 or JTT-130.
  • the compounds according to the invention are administered in combination with a PPAR-gamma agonist such as, by way of example and preferably, pioglitazonc or rosiglitazonc.
  • a PPAR-gamma agonist such as, by way of example and preferably, pioglitazonc or rosiglitazonc.
  • the compounds according to the invention are administered in combination with a PPAR-delta agonist such as, for example and preferably, GW 501516 or BAY 68-5042.
  • a PPAR-delta agonist such as, for example and preferably, GW 501516 or BAY 68-5042.
  • the compounds according to the invention are administered in combination with a cholesterol absorption inhibitor such as, by way of example and preferably, ezetimibe, tiqueside or pamaqueside.
  • a cholesterol absorption inhibitor such as, by way of example and preferably, ezetimibe, tiqueside or pamaqueside.
  • the compounds according to the invention are administered in combination with a lipase inhibitor, a preferred example being orlistat.
  • the compounds according to the invention are administered in combination with a polymeric bile acid adsorbent such as, by way of example and preferably, cholestyramine, colestipol, colesolvam, CholestaGel or colestimide.
  • a polymeric bile acid adsorbent such as, by way of example and preferably, cholestyramine, colestipol, colesolvam, CholestaGel or colestimide.
  • the compounds according to the invention are administered in combination with an lipoprotein(a) antagonist such as, by way of example and preferably, gemcabene calcium (CI-1027) or nicotinic acid.
  • an lipoprotein(a) antagonist such as, by way of example and preferably, gemcabene calcium (CI-1027) or nicotinic acid.
  • the present invention further provides medicaments which comprise at least one compound according to the invention, typically together with one or more inert, nontoxic, pharmaceutically suitable auxiliaries, and the use thereof for the aforementioned purposes.
  • the compounds according to the invention may act systemically and/or locally.
  • they can be administered in a suitable manner, for example by the oral, parenteral, pulmonal, nasal, sublingual, lingual, buccal, rectal, dermal, transdermal, conjunctival, otic route, or as an implant or stent.
  • the compounds according to the invention can be administered in administration forms suitable for these administration routes.
  • Suitable administration forms for oral administration are those which work according to the prior art, which release the compounds according to the invention rapidly and/or in a modified manner and which contain the compounds according to the invention in crystalline and/or amorphized and/or dissolved form, for example tablets (uncoated or coated tablets, for example with gastric juice-resistant or retarded-dissolution or insoluble coatings which control the release of the compound according to the invention), tablets or films/wafers which disintegrate rapidly in the oral cavity, films/lyophilizates or capsules (for example hard or soft gelatin capsules), sugar-coated tablets, granules, pellets, powders, emulsions, suspensions, aerosols or solutions.
  • tablets uncoated or coated tablets, for example with gastric juice-resistant or retarded-dissolution or insoluble coatings which control the release of the compound according to the invention
  • tablets or films/wafers which disintegrate rapidly in the oral cavity
  • films/lyophilizates or capsules for example hard or soft gelatin
  • Parenteral administration can bypass an absorption step (e.g. intravenously, intraarterially, intracardially, intraspinally or intralumbally) or include an absorption (e.g. intramuscularly, subcutaneously, intracutaneously, percutaneously or intraperitoneally).
  • Administration forms suitable for parenteral administration include preparations for injection and infusion in the form of solutions, suspensions, emulsions, lyophilizates or sterile powders.
  • suitable examples are inhalable medicament forms (including powder inhalers, nebulizers), nasal drops, solutions or sprays, tablets, films/wafers or capsules for lingual, sublingual or buccal administration, suppositories, car or eye preparations, vaginal capsules, aqueous suspensions (lotions, shaking mixtures), lipophilic suspensions, ointments, creams, transdermal therapeutic systems (e.g. patches), milk, pastes, foams, sprinkling powders, implants or stents.
  • Oral or parenteral administration is preferred, especially oral administration.
  • the compounds according to the invention can be converted to the administration forms mentioned. This can be done in a manner known per se, by mixing with inert, nontoxic, pharmaceutically suitable excipients.
  • excipients include carriers (for example microcrystalline cellulose, lactose, mannitol), solvents (e.g. liquid polyethylene glycols), emulsifiers and dispersing or wetting agents (for example sodium dodecylsulphate, polyoxysorbitan oleate), binders (for example polyvinylpyrrolidone), synthetic and natural polymers (for example albumin), stabilizers (e.g. antioxidants, for example ascorbic acid), dyes (e.g. inorganic pigments, for example iron oxides) and flavour and/or odour correctants.
  • carriers for example microcrystalline cellulose, lactose, mannitol
  • solvents e.g. liquid polyethylene glycols
  • emulsifiers and dispersing or wetting agents for example sodium do
  • the dosage is about 0.01 to 100 mg/kg, preferably about 0.01 to 20 mg/kg and most preferably 0.1 to 10 mg/kg of body weight.
  • Method 1 MS instrument type: Waters ZQ; apparatus type HPLC: Agilent 1100 Series; UV DAD; column: Thermo Hypersil GOLD 3 ⁇ 20 mm ⁇ 4 mm; mobile phase A: 1 l of water+0.5 ml of 50% strength formic acid, mobile phase B: 1 l of acetonitrile+0.5 ml of 50% strength formic acid; gradient: 0.0 min 100% A 3.0 min 10% A 4.0 min 10% A 4.1 min 100% A (flow rate 2.5 ml/min); oven: 55° C.; flow rate: 2 ml/min; UV detection: 210 nm.
  • Method 2 Instrument: Waters ACQUITY SQD UPLC System; column: Waters Acquity UPLC HSS T3 1.8 ⁇ 50 ⁇ 1 mm; mobile phase A: 1 l of water+0.25 ml of 99% strength formic acid, mobile phase B: 1 l of acetonitrile+0.25 ml of 99% strength formic acid; gradient: 0.0 min 90% A ⁇ 1.2 min 5% A ⁇ 2.0 min 5% A oven: 50° C.; flow rate: 0.40 ml/min; UV detection: 210-400 nm.
  • Method 3 Instrument: Micromass Quattro Premier with Waters UPLC Acquity; column: Thermo Hypersil GOLD 1.9 ⁇ 50 mm ⁇ 1 mm; mobile phase A: 1 l of water+0.5 ml of 50% strength formic acid, mobile phase B: 1 l of acetonitrile+0.5 ml of 50% strength formic acid; gradient: 0.0 min 90% A 0.1 min 90% A 1.5 min 10% A 2.2 min 10% A; oven 50° C.; flow rate: 0.33 ml/min;
  • UV detection 210 nm.
  • PLM The polarized light microscopy was carried out using a Clemex PS3 polarized light microscope particle size system with a Leica DM microscope procided with 50 ⁇ , 100 ⁇ , 200 ⁇ , and 500 ⁇ lenses, a high-resolution monochrome 1600 ⁇ 1200 pixel digital camera and a motorized X-Y Marzhauser station (controlled by a Clemex ST-2000 controller).
  • the samples of the crystalline material were measured on a glass slide (76 ⁇ 26 mm) in a drop of oil, the sample being covered with a cover glass (22 ⁇ 40 mm).
  • the melting points were determined by dynamic differential calorimetry. The determination was carried out using a Mettler-Toledo 823e DSC instrument provided with a TSO801RO sample robot and STAR e software. About 1.5 to 3 mg of the sample were weighed out into a small aluminium pan, which was then closed with a perforated cap. The heat flow was measured in a temperature range of from 30 to 400° C. at a heating rate of 10° C./min and under an argon stream of 30 ml/min.
  • thermogravimetric analysis was carried out using a Mettler-Toledo TGA/SDTA851 e TGA instrument provided with a TSO801RO sample robot and STAR e software. About 1.5 to 3 mg of the sample were weighed out into a small open aluminium pan (100 The sample weight was measured in a temperature range of from 30 to 400° C. at a heating rate of 10° C./min and under an argon stream of 30 ml/min.
  • reaction mixture was filtered with suction through kieselguhr and the filter product was washed three times with ethyl acetate (517 ml each time).
  • the organic phase was separated off and the aqueous phase was washed with ethyl acetate (258 ml).
  • the combined organic phases were washed once with saturated aqueous sodium hydrogen carbonate solution (414 ml), dried and concentrated under reduced pressure.
  • Dichloromethane (388 ml) was added to the crystals obtained in this manner, and the mixture was stirred for 20 min. The mixture was once more filtered off with suction, washed with diethyl ether and sucked dry.
  • the diazonium salt thus prepared was added to a cold (0° C.) solution of 12.81 g (85.45 mmol) of sodium iodide in acetone (329 ml), and the mixture was stirred at RT for 30 min.
  • the reaction mixture was poured into ice-water (1.8 l) and extracted twice with ethyl acetate (487 ml each time).
  • the collected organic phases were washed with saturated aqueous sodium chloride solution (244 ml), dried, filtered and concentrated. This gave 12.1 g (86% purity, 60% of theory) of the desired compound in the form of a brown solid.
  • the crude product was reacted without further purification.
  • the aqueous phase was separated off and extracted two more times with ethyl acetate (200 ml each time).
  • the combined organic phases were washed twice with 10% strength aqueous sodium chloride solution (100 ml each time), dried and concentrated under reduced pressure.
  • the crude product was reacted without further purification.
  • the mixture was then concentrated to dryness and the residue was taken up in water (100 ml) and ethyl acetate (100 ml) and adjusted to a pH of 10 using 2N aqueous sodium hydroxide solution.
  • the mixture was stirred intensively at RT for about 1 h.
  • the resulting suspension was filtered with suction and the filter product was washed with ethyl acetate (100 ml), with water (100 ml) and once more with ethyl acetate (100 ml).
  • the residue was dried under high vacuum over phosphorus pentoxide.
  • a quantity of 3.470 g (8.138 mmol) of the compound from Example 1 was suspended in 35 ml of THF, admixed at 0° C. with 358 mg (8.952 mmol) of sodium hydride (60% suspension in mineral oil) and stirred at 0° C. for 90 min, in the course of which a solution was formed.
  • a quantity of 2.519 g (8.952 mmol) of 2,2,2-trifluoroethyl trichloromethanesulphonate was added and the mixture was stirred at RT for 48 h. It was then stirred with water and concentrated on a rotary evaporator.
  • Example 1 A solution of 100 mg (0.235 mmol) of Example 1 in 2 ml of 1,4-dioxane was prepared in a brown 5 ml glass bottle. In succession, 2 ml of isopropanol and 235 ⁇ l (0.235 mmol) of 1M hydrochloric acid were added to this solution, and the solution was stirred at RT until the solvents had evaporated. Air-drying gave 102 mg (94% of theory) of the title compound.
  • TGA 1% weight loss at 80° C.
  • Example 1 A solution of 100 mg (0.235 mmol) of Example 1 in 2 ml of 1,4-dioxane was prepared in a brown 5 ml glass bottle. In succession, 2 ml of isopropanol and a solution of 938 ⁇ l (0.235 mmol) of 0.25M sulphuric acid were added to this solution, and the solution was stirred at RT until the solvents had evaporated. Air-drying gave 103 mg (83.7% of theory) of the title compound.
  • Example 1 A solution of 100 mg (0.235 mmol) of Example 1 in 2 ml of 1,4-dioxane was prepared in a brown 5 ml glass bottle. In succession, 2 ml of THF and a solution of 16 ⁇ l (0.235 mmol) of 85% strength phosphoric acid in 0.3 ml of water were added to this solution, and the solution was stirred at RT until the solvents had evaporated. Air-drying gave 105 mg (85.4% of theory) of the title compound.
  • Example 1 A solution of 100 mg (0.235 mmol) of Example 1 in 2 ml of 1,4-dioxane was prepared in a brown 5 ml glass bottle. In succession, 2 ml of ethanol and a solution of 22.5 mg (0.235 mmol) of methanesulphonic acid in 0.3 ml of water were added to this solution, and the solution was stirred at RT until the solvents had evaporated. Air-drying gave 103 mg (84% of theory) of the title compound.
  • Example 1 A solution of 100 mg (0.235 mmol) of Example 1 in 2 ml of 1,4-dioxane was prepared in a brown 5 ml glass bottle. In succession, 2 ml of isopropanol and 44.6 mg (0.235 mmol) of ethane-1,2-disulphonic acid were added to this solution, and the solution was stirred at RT until the solvents had evaporated. Air-drying gave 111 mg (73.7% of theory) of the title compound.
  • Example 1 A solution of 100 mg (0.235 mmol) of Example 1 in 2 ml of 1,4-dioxane was prepared in a brown 5 ml glass bottle. In succession, 2 ml of isopropanol and 27.2 mg (0.235 mmol) of maleic acid were added to this solution, and the solution was stirred at RT until the solvents had evaporated. Air-drying gave 108 mg (84.9% of theory) of the title compound.
  • Example 1 A solution of 100 mg (0.235 mmol) of Example 1 in 2 ml of isopropanol was prepared in a brown 5 ml glass bottle. In succession, 2 ml of isopropanol and 0.235 ⁇ l (0.235 mmol) of 1M nitric acid were added to this solution, and the solution was stirred at RT until the solvents had evaporated. Air-drying gave 103 mg (89.7% of theory) of the title compound.
  • Rabbits are stunned by a blow to the neck and exsanguinated.
  • the aorta is removed, freed from adhering tissue and divided into rings of a width of 1.5 mm.
  • the rings are placed individually under an initial tension in 5 ml organ baths with Krebs-Henseleit solution which is at 37° C., is gassed with carbogen and has the following composition (in each case mM): sodium chloride 119; potassium chloride: 4.8; calcium chloride dihydrate: 1; magnesium sulphate heptahydrate: 1.4; potassium dihydrogenphosphate: 1.2; sodium bicarbonate: 25; glucose: 10.
  • the force of contraction is detected with Statham UC2 cells, amplified and digitized via A/D converters (DAS-1802 HC, Keithley Instruments, Kunststoff) and recorded in parallel on chart recorders.
  • A/D converters DAS-1802 HC, Keithley Instruments, Kunststoff
  • phenylephrine is added to the bath cumulatively in increasing concentration.
  • the substance to be investigated is added in each further run in increasing dosage in each case, and the height of the contraction achieved is compared with the height of the contraction reached in the last preceding run.
  • the concentration necessary to reduce the height of the control value by 50% is calculated from this (IC 50 value).
  • the standard administration volume is 5 ⁇ l and the proportion of DMSO in the bath solution corresponds to 0.1%.
  • the cellular activity of the compounds according to the invention is determined using a recombinant guanylate cyclase reporter cell line, as described in F. Wunder et al., Anal. Biochem. 339, 104-112 (2005).
  • the system consists of 3 main components:
  • the telemetry system makes it possible to continuously record blood pressure, heart rate and body motions of conscious animals in their usual habitat.
  • the experimental animals After transmitter implantation, the experimental animals are housed singly in type 3 Makrolon cages. They have free access to standard feed and water.
  • the day/night rhythm in the experimental laboratory is changed by the room lighting at 6.00 am and at 7.00 pm.
  • the telemetry transmitters TA11 PA-C40 used are surgically implanted under aseptic conditions in the experimental animals at least 14 days before the first experimental use.
  • the animals instrumented in this way can be employed repeatedly after the wound has healed and the implant has settled.
  • the fasted animals are anaesthetized with pentobarbital (Nembutal, Sanofi: 50 mg/kg i.p.) and shaved and disinfected over a large area of their abdomens.
  • pentobarbital Nembutal, Sanofi: 50 mg/kg i.p.
  • the liquid-filled measuring catheter of the system is inserted into the descending aorta in the cranial direction above the bifurcation and fixed with tissue glue (VetBonDTM, 3M).
  • the transmitter housing is fixed intraperitoneally to the abdominal wall muscle, and layered closure of the wound is performed.
  • An antibiotic (Tardomyocel COMP, Bayer, 1 ml/kg s.c.) is administered postoperatively for prophylaxis of infection.
  • the test substances are dissolved in suitable solvent mixtures, or suspended in 0.5% strength Tylose, appropriate for an administration volume of 5 ml/kg of body weight.
  • a solvent-treated group of animals is employed as control.
  • the telemetry measuring unit present is configured for 24 animals. Each experiment is recorded under an experiment number (Vyear month day).
  • Each of the instrumented rats living in the system is assigned a separate receiving antenna (1010 Receiver, DSI).
  • the implanted transmitters can be activated externally by means of an incorporated magnetic switch and are switched to transmission in the run-up to the experiment.
  • the emitted signals can be detected online by a data acquisition system (DataquestTM A.R.T. for Windows, DSI) and be appropriately processed.
  • the data are stored in each case in a file created for this purpose and bearing the experiment number.
  • the acquisition of measured values is repeated under computer control at 5-minute intervals.
  • the source data obtained as absolute value are corrected in the diagram with the currently measured barometric pressure (Ambient Pressure Reference Monitor; APR-1) and stored as individual data. Further technical details are given in the extensive documentation from the manufacturing company (DSI).
  • test substances are administered at 9.00 am on the day of the experiment. Following the administration, the parameters described above are measured over 24 hours.
  • the acquired individual data are sorted using the analysis software (DataquestTM A.R.T.TM Analysis).
  • the blank value is assumed to be the time 2 hours before administration of the substance, so that the selected data set includes the period from 7.00 am on the day of the experiment to 9.00 am on the following day.
  • the data are smoothed over a presettable time by determination of the average (15-minute average) and transferred as a text file to a storage medium.
  • the measured values presorted and compressed in this way are transferred into Excel templates and tabulated.
  • the data obtained are stored in a dedicated file carrying the number of the experiment. Results and test protocols are filed in paper form sorted by numbers.
  • Example 2 Dosage Dosage 0.3 mg/kg 3.0 mg/kg 0.3 mg/kg Vehicle p.o. p.o. Vehicle p.o. hours mean mean mean mean hours mean mean after blood blood blood after blood blood substance pressure pressure pressure substance pressure pressure administration (mm Hg) (mm Hg) administration (mm Hg) (mm Hg) 0 153.6 151.0 149.0 0 149.0 161.3 1 164.5 148.4 129.3 1 158.2 145.7 2 146.7 136.4 111.1 2 142.2 130.5 3 145.4 130.6 106.0 3 149.2 121.5 4 149.6 129.1 107.8 4 152.3 123.1 5 149.9 132.8 109.3 5 155.8 121.6 6 151.6 125.6 106.8 6 147.3 123.8 7 147.6 131.9 110.9 7 147.3 124.4 8 147.5 131.8 109.8 8 149.3 128.7 9 150.8 138.5 114.3 9 151.0 133.7 10
  • the pharmacokinetic parameters of the substance are determined in male CD-1 mice, male Wistar rats and female beagles.
  • the administration volume is 10 ml/kg for mice, 5 ml/kg for rats and 0.5 ml/kg for dogs.
  • Intravenous administration is via a formulation of species-specific plasma/DMSO (99/1) in the case of mice and rats and via water/PEG400/ethanol (50/40/10) in the case of dogs.
  • a silicone catheter is inserted into the right Vena jugularis externa of the rats before the administration of substance.
  • Substance administration is as i.v. bolus in the case of mice, as i.v. bolus or via a 15-minute infusion in the case of rats and via a 15-minute infusion in the case of dogs.
  • Removal of blood is after 0.033, 0.083, 0.17, 0.5, 1, 2, 3, 4, 6, 7 and 24 hours in the case of mice and, after a 15-minute infusion, after 0.083, 0.25, 0.28 0.33, 0.42, 0.75, 1, 2, 3, 4, 6, 7 and 24 hours in the case of dogs and rats and after an i.v. bolus administration, after 0.033, 0.083, 0.17, 0.5, 1, 2, 3, 4, 6, 7 and 24 hours in the case of rats.
  • oral administration of the dissolved substance via gavage is carried out based on a water/PEG400/ethanol formulation (50/40/10).
  • the removal of blood from rats and dogs is after 0.083, 0.17, 0.5, 0.75, 1, 2, 3, 4, 6, 7 and 24 hours.
  • the blood is removed into heparinized tubes.
  • the blood plasma is then obtained by centrifugation; if required, it can be stored at ⁇ 20° C. until further processing.
  • Example 1 a binary mobile phase gradient at 400 ⁇ l/min is used (A: 0.01M ammonium acetate buffer pH 6.8, B: 0.1% formic acid in acetonitrile): 0 min (90% A), 1 min (90% A), 3.5 min (15% A), 4.5 min (15% A), 4.6 min (90% A), 7 min (90% A).
  • Example 2 a binary mobile phase gradient at 500 ⁇ l/min is used instead (A: 0.01M ammonium acetate buffer pH 3.0, B: 0.1% formic acid in acetonitrile): 0 min (90% A), 1.5 min (90% A), 3.5 min (10% A), 4.5 min (10% A), 5 min (90% A), 7 min (90% A).
  • Example 3 a binary mobile solvent gradient at 500 ⁇ l/min is used instead (A: 0.01M ammonium acetate buffer pH 6.8, B: 0.1% formic acid in acetonitrile): 0 min (90% A), 1 min (90% A), 3 min (10% A), 4 min (10% A), 4.5 min (90% A), 6 min (90% A).
  • the temperature of the Turbo V ion source is 500° C.
  • Example 2 The following MS instrument parameters are used: curtain gas 20 units (Example 1), 16 units (Example 2) or 15 units (Example 3), ion spray voltage 5 kV (Example 1/2) or 4.5 units (Example 3), gas 1 35 units (Example 1/3) or 25 units (Example 2), gas 2 30 units, CAD gas 4 units (Example 1/3) or 3 units (Example 2).
  • the substances are quantified by peak heights or areas using extracted ion chromatograms of specific MRM experiments.
  • the plasma concentration/time plots determined are used to calculate the pharmacokinetic parameters such as AUC, C max , MRT (mean residence time), t 1/2 (half life) and CL (clearance) employing the validated pharmacokinetic calculation programs KinEx (Vers. 2.5 and 3).
  • the substance quantification is carried out in plasma, it is necessary to determine the blood/plasma distribution of the substance to be able to adjust the pharamacokinetic parameters in an appropriate manner.
  • a defined amount of substance is incubated in heparinized whole blood of the species in question in a rocking roller mixer for 20 min. After centrifugation at 1000 g, the plasma concentration is measured (see above) and determined by calculating the quotient of the c b /c p values.
  • the substances according to the invention show a surprisingly favourable safety profile in vivo which was established by non-clinical safety studies according to OECD (OECD guidelines for testing of chemicals, No. 407) and ICH (3BS2A) guidelines.
  • the compounds according to the invention can be converted to pharmaceutical formulations as follows
  • the mixture of the compound according to the invention, lactose and starch is granulated with a 5% solution (w/w) of the PVP in water. After drying, the granules are mixed with the magnesium stearate for 5 minutes. This mixture is pressed with a conventional tableting press (for tablet dimensions see above).
  • the guide value used for the pressing is a pressing force of 15 kN.
  • a single dose of 100 mg of the compound according to the invention corresponds to 10 ml of oral suspension.
  • Rhodigel is suspended in ethanol and the compound according to the invention is added to the suspension.
  • the water is added while stirring.
  • the mixture is stirred for approx. 6 h until swelling of the Rhodigel has ended.
  • a single dose of 100 mg of the compound according to the invention corresponds to 20 g of oral solution.
  • the compound according to the invention is suspended in the mixture of polyethylene glycol and polysorbate while stirring. The stirring operation is continued until dissolution of the compound according to the invention is complete.
  • the compound according to the invention is dissolved in a concentration below the saturation solubility in a physiologically acceptable solvent (e.g. isotonic saline, glucose solution 5% and/or PEG 400 solution 30%).
  • a physiologically acceptable solvent e.g. isotonic saline, glucose solution 5% and/or PEG 400 solution 30%.
  • the solution is subjected to sterile filtration and dispensed into sterile and pyrogen-free injection vessels.

Abstract

The present application relates to novel substituted 5-fluoro-1H-pyrazolopyridines, to processes for their preparation, to their use alone or in combinations for the treatment and/or prophylaxis of diseases, and to their use for producing medicaments for the treatment and/or prophylaxis of diseases, in particular for the treatment and/or prophylaxis of cardiovascular disorders.

Description

This application is a divisional of U.S. application Ser. No. 15/981,598, filed on May 16, 2018, now U.S. Pat. No. 10,736,896, which is a continuation of U.S. application Ser. No. 15/348,545, filed on Nov. 10, 2016, now U.S. Pat. No. 9,993,476, which is a continuation of U.S. application Ser. No. 14/996,678, filed on Jan. 15, 2016, which is a continuation of U.S. application Ser. No. 14/552,122, filed on Nov. 24, 2014, now U.S. Pat. No. 9,266,885, which is a continuation of U.S. application Ser. No. 13/851,373 filed on Mar. 27, 2013, now U.S. Pat. No. 8,921,377, which is a continuation of U.S. application Ser. No. 13/111,856, filed on May 19, 2011, now U.S. Pat. No. 8,420,656, and claims priority under 35 U.S.C. § 119 to German Patent Application No. 102010021637.2, filed May 26, 2010.
The present application relates to novel substituted 5-fluoro-1H-pyrazolopyridines, to processes for their preparation, to their use alone or in combinations for the treatment and/or prophylaxis of diseases, and to their use for producing medicaments for the treatment and/or prophylaxis of diseases, in particular for the treatment and/or prophylaxis of cardiovascular disorders.
One of the most important cellular transmission systems in mammalian cells is cyclic guanosine monophosphate (cGMP). Together with nitrogen monoxide (NO), which is released from the endothelium and transmits hormonal and mechanical signals, it forms the NO/cGMP system. Guanylate cyclases catalyze the biosynthesis of cGMP from guanosine triphosphate (GTP). The known representatives of this family can be classified both according to structural features and according to the type of ligands into two groups: the particulate guanylate cyclases which can be stimulated by natriuretic peptides, and the soluble guanylate cyclases which can be stimulated by NO. The soluble guanylate cyclases consist of two subunits and very probably contain one haem per heterodimer, which is part of the regulatory site. It is of central importance for the activation mechanism. NO is able to bind to the iron atom of haem and thus markedly increase the activity of the enzyme. Haem-free preparations cannot, by contrast, be stimulated by NO. Carbon monoxide (CO) is also able to attach to the central iron atom of haem, but the stimulation by CO is distinctly less than that by NO.
By forming cGMP, and owing to the resulting regulation of phosphodiesterases, ion channels and protein kinases, guanylate cyclase plays an important role in various physiological processes, in particular in the relaxation and proliferation of smooth muscle cells, in platelet aggregation and platelet adhesion and in neuronal signal transmission, and also in disorders which are based on a disturbance of the abovementioned processes. Under pathophysiological conditions, the NO/cGMP system may be suppressed, which may lead for example to high blood pressure, platelet activation, increased cellular proliferation, endothelial dysfunction, atherosclerosis, angina pectoris, heart failure, myocardial infarction, thromboses, stroke and sexual dysfunction.
Owing to the expected high efficiency and few side effects, a treatment of such disorders which targets the influence of the cGMP signal path in organisms and is NO-independent is a promising approach.
Hitherto, for the therapeutic stimulation of the soluble guanylate cyclase use has exclusively been made of compounds such as organic nitrates whose effect is based on NO. This is formed by bioconversion and activates soluble guanylate cyclase by attack at the central iron atom of haem. In addition to the side effects, the development of tolerance is one of the decisive disadvantages of this type of treatment.
In recent years, some substances have been described which stimulate soluble guanylate cyclase directly, i.e. without prior release of NO, such as, for example 3-(5′-hydroxymethyl-2′-furyl)-1-benzylindazole [YC-1; Wu et al., Blood 84 (1994), 4226; Mülsch et al., Brit. J. Pharmacol. 120 (1997), 681], fatty acids [Goldberg et al., J. Biol. Chem. 252 (1977), 1279], diphenyliodonium hexafluorophosphate [Pettibone et al., Eur. J. Pharmacol. 116 (1985), 307], isoliquiritigenin [Yu et al., Brit. J. Pharmacol. 114 (1995), 1587] and various substituted pyrazole derivatives (WO 98/16223).
WO 00/06569 discloses fused pyrazole derivatives and WO 03/095451 carbamate-substituted 3-pyrimidinylyrazolopyridines as stimulators of soluble guanylate cyclase.
It is an object of the present invention to provide novel substances which act as stimulators of soluble guanylate cyclase and which have an identical or improved therapeutic profile compared to compounds known from the prior art, such as, for example, with respect to their in vivo properties such as, for example, their pharmacokinetic and pharmacodynamic behaviour and/or their dose-activity relationship.
The present invention provides compounds of the general formula (I)
Figure US11439642-20220913-C00001

in which
  • R1 represents hydrogen or (C1-C4)-alkyl,
    • where (C1-C4)-alkyl may be substituted by one or two substituents independently of one another selected from the group consisting of fluorine and trifluoromethyl,
      and their N-oxides, salts, solvates, salts of the N-oxides and solvates of the N-oxides and salts.
Compounds according to the invention are the compounds of the formula (I) and the N-oxides, salts, solvates and solvates of the N-oxides and salts thereof, the compounds, encompassed by formula (I), of the formulae specified hereinafter and the N-oxides, salts, solvates and solvates of the N-oxides and salts thereof, and the compounds encompassed by formula (I) and specified hereinafter as working examples and the N-oxides, salts, solvates and solvates of the N-oxides and salts thereof, to the extent that the compounds encompassed by formula (I) and specified hereinafter are not already N-oxides, salts, solvates and solvates of the N-oxides and salts.
In the context of the present invention, preferred salts are physiologically acceptable salts of the compounds according to the invention. Also encompassed are salts which are not themselves suitable for pharmaceutical applications but can be used, for example, for isolation or purification of the compounds according to the invention.
Physiologically acceptable salts of the compounds according to the invention include acid addition salts of mineral acids, carboxylic acids and sulphonic acids, for example salts of hydrochloric acid, hydrobromic acid, sulphuric acid, phosphoric acid, methanesulphonic acid, ethanesulphonic acid, toluenesulphonic acid, benzenesulphonic acid, naphthalenedisulphonic acid, formic acid, acetic acid, trifluoroacetic acid, propionic acid, lactic acid, tartaric acid, malic acid, citric acid, fumaric acid, maleic acid and benzoic acid.
Physiologically acceptable salts of the compounds according to the invention also include salts of conventional bases, by way of example and with preference alkali metal salts (e.g. sodium and potassium salts), alkaline earth metal salts (e.g. calcium and magnesium salts) and ammonium salts derived from ammonia or organic amines having 1 to 16 carbon atoms, by way of example and with preference ethylamine, diethylamine, triethylamine, ethyldiisopropylamine, monoethanolamine, diethanolamine, triethanolamine, dicyclohexylamine, dimethylaminoethanol, procaine, dibenzylamine, N-methylmorpholine, arginine, lysine, ethylenediamine and N-methylpiperidine.
In the context of the invention, solvates refer to those forms of the compounds according to the invention which, in the solid or liquid state, form a complex by coordination with solvent molecules. Hydrates are a specific form of the solvates in which the coordination is with water. Preferred solvates in the context of the present invention are hydrates.
Depending on their structure, the compounds according to the invention may exist in different stereoisomeric forms, i.e. in the form of configurational isomers or if appropriate also as conformational isomers (enantiomers and/or diastereomers, including those in the case of atropisomers). The present invention therefore encompasses the enantiomers or diastereomers and the respective mixtures thereof. The stereoisomerically uniform constituents can be isolated from such mixtures of enantiomers and/or diastereomers in a known manner; chromatography processes are preferably used for this, in particular HPLC chromatography on an achiral or chiral phase.
Where the compounds according to the invention can occur in tautomeric forms, the present invention encompasses all the tautomeric forms.
The present invention also encompasses all suitable isotopic variants of the compounds according to the invention. An isotopic variant of a compound according to the invention is understood here to mean a compound in which at least one atom within the compound according to the invention compound has been exchanged for another atom of the same atomic number, but with a different atomic mass than the atomic mass which usually or predominantly occurs in nature. Examples of isotopes which can be incorporated into a compound according to the invention are those of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulphur, fluorine, chlorine, bromine and iodine, such as 2H (deuterium), 3H (tritium), 13c, 14C, 15N, 17O, 18O, 32P, 33P, 33S, 34S, 35S, 36S, 18F, 36Cl, 82Br, 123I, 124I, 129I and 131I. Particular isotopic variants of a compound according to the invention, especially those in which one or more radioactive isotopes have been incorporated, may be beneficial, for example, for the examination of the mechanism of action or of the active compound distribution in the body; due to comparatively easy preparability and detectability, especially compounds labelled with 3H or 14C isotopes are suitable for this purpose. In addition, the incorporation of isotopes, for example of deuterium, can lead to particular therapeutic benefits as a consequence of greater metabolic stability of the compound, for example an extension of the half-life in the body or a reduction in the active dose required; such modifications of the compounds according to the invention may therefore in some cases also constitute a preferred embodiment of the present invention. Isotopic variants of the compounds according to the invention can be prepared by processes known to those skilled in the art, for example by the methods described below and the methods described in the working examples, by using corresponding isotopic modifications of the particular reagents and/or starting compounds therein.
Moreover, the present invention also encompasses prodrugs of the compounds according to the invention. Here, the term “prodrugs” refers to compounds which for their part can be biologically active or inactive, but are converted (for example metabolically or hydrolytically) into compounds according to the invention during their dwell time in the body.
In the context of the present invention, unless specified otherwise, the substituents are defined as follows:
In the context of the invention, alkyl represents a straight-chain or branched alkyl radical having 1 to 4 carbon atoms. The following may be mentioned by way of example and by way of preference: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, 1-methylpropyl, tert-butyl.
In the context of the invention, halogen represents fluorine, chlorine, bromine and iodine.
If radicals in the compounds according to the invention are substituted, the radicals may be mono- or polysubstituted, unless specified otherwise. In the context of the present invention, all radicals which occur more than once are defined independently of one another. Substitution by one, two or three identical or different substituents is preferred.
In the context of the present invention, preference is given to compounds of the formula (I) in which
  • R1 represents hydrogen or methyl,
where methyl may be substituted by a trifluoromethyl substituent, and their salts, solvates and solvates of the salts.
In the context of the present invention, particular preference is given to the following compounds of the formula (I):
  • methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate
  • methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}methylcarbamate
  • methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}(2,2,2-trifluoroethyl)carbamate
  • methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate hydrochloride
  • methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate sulphate
  • methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate phosphate
  • methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate mesylate
  • methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate ethane-1,2-disulphonate
  • methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate maleate
  • methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate nitrate
The individual radical definitions specified in the respective combinations or preferred combinations of radicals are, independently of the respective combinations of the radicals specified, also replaced as desired by radical definitions of other combinations.
Particular preference is given to combinations of two or more of the preferred ranges mentioned above.
The invention further provides a process for preparing the compounds of the formula (I) according to the invention, characterized in that the compound of the formula (II)
Figure US11439642-20220913-C00002
  • [A] is reacted in an inert solvent in the presence of hexabutyltin and a suitable palladium catalyst with intermediate formation of a tin species with the compound of the formula (III)
Figure US11439642-20220913-C00003
    • to give the compound of the formula (IV)
Figure US11439642-20220913-C00004
    • this is then reduced in an inert solvent with a suitable reducing agent to give the compound of the formula (V)
Figure US11439642-20220913-C00005
    • and this is then reacted in the presence of a suitable base in the presence or absence of a solvent with methyl chloroformate to give the compound of the formula (I-A)
Figure US11439642-20220913-C00006

or
  • [B] the compound of the formula (II) is reacted in an inert solvent with copper cyanide to give the compound of the formula (VI)
Figure US11439642-20220913-C00007
    • this is then, under acidic conditions, converted into the compound of the formula (VII)
Figure US11439642-20220913-C00008
    • this is subsequently reacted in an inert solvent in the presence of a suitable base with the compound of the formula (VIII)
Figure US11439642-20220913-C00009
    • to give the compound of the formula (IX)
Figure US11439642-20220913-C00010
    • and this is then reduced in an inert solvent in the presence of a suitable reducing agent to give the compound (V), and this is subsequently reacted further according to process [A] to give compound (I-A),
      or
  • [C] the compound of the formula (I-A) is reacted in an inert solvent in the presence of a suitable base with a compound of the formula (X)
    R1A—X1  (X),
    • R1A represents (C1-C4)-alkyl,
      • where (C1-C4)-alkyl may be substituted by one or two substituents independently of one another selected from the group consisting of fluorine and trifluoromethyl,
    • and
    • X1 represents a leaving group such as, for example, halogen, in particular bromine or iodine, trichloromethanesulphonate, mesylate or tosylate
    • to give a compound of the formula (I-B)
Figure US11439642-20220913-C00011
    • in which R1A has the meaning given above,
      and the resulting compounds of the formulae (I-A) and (I-B) are, where appropriate, converted with the appropriate (i) solvents and/or (ii) acids or bases into their solvates, salts and/or solvates of the salts.
Inert solvents for process step (II)+(III)→(IV) are, for example, alcohols such as methanol, ethanol, n-propanol, isopropanol, n-butanol or tert-butanol, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, hydrocarbons such as benzene, xylene, toluene, hexane, cyclohexane or mineral oil fractions, or other solvents such as dimethylformamide (DMF), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU), dimethylacetamide, N-methylpyrrolidone (NMP), pyridine, acetonitrile, sulpholane or else water. It is equally possible to use mixtures of the solvents mentioned. Preference is given to dioxane.
Suitable palladium catalysts for process step (II)+(III)→(IV) are, for example, palladium on activated carbon, palladium(II) acetate, tetrakis(triphenylphosphine)palladium(0), bis(triphenylphosphine)palladium(II) chloride, bis(acetonitrile)palladium(II) chloride and [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II)/dichloromethane complex, if appropriate in combination with additional phosphane ligands such as, for example, (2-biphenyl)di-tert-butylphosphine, dicyclohexyl[2′,4′,6′-tris(1-methylethyl)biphenyl-2-yl]phosphane (XPHOS), bis(2-phenylphosphinophenyl) ether (DPEphos) or 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene (Xantphos) [cf., for example, Hassan J. et al., Chem. Rev. 102, 1359-1469 (2002)]. Preference is given to using tetrakis(triphenylphosphine)palladium(0).
The reaction (II)+(III)→(IV) is generally carried out in a temperature range of from +20° C. to +180° C., prepferably from +50° C. to +120° C., if appropriate in a microwave. The reaction can be carried out at atmospheric, elevated or reduced pressure (for example from 0.5 to 5 bar). The reaction is generally carried out at atmospheric pressure.
The reductions (IV)→(V) and (IX)→(V) are carried out in the presence of a suitable catalyst in an inert solvent in a temperature range of from +20° C. to +40° C. under hydrogen of atmospheric pressure.
Inert solvents for the reductions (IV)→(V) and (IX)→(V) are, for example, alcohols such as methanol, ethanol, n-propanol, isopropanol, n-butanol or tert-butanol, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, or other solvents such as dimethylformamide (DMF), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU), N-methylpyrrolidone (NMP), pyridine, acetonitrile or else water. It is equally possible to use mixtures of the solvents mentioned. Preference is given to DMF and pyridine.
Suitable catalysts for the reactions (IV)→(V) and (IX)→(V) are, for example, palladium on activated carbon, platinum on carbon, palladium hydroxide or Raney nickel.
Alternatively, the reductions (IV)→(V) and (IX)→(V) can be carried out using a metal or metal salt such as, for example, iron, zinc or tin(II) chloride in a suitable acid such as, for example, hydrogen chloride/hydrochloric acid, sulphuric acid, phosphoric acid or acetic acid in a temperature range of from +20° C. to +140° C.
Inert solvents for process step (V)→(I-A) are, for example, alcohols such as methanol, ethanol, n-propanol, isopropanol, n-butanol or tert-butanol, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, trichloroethylene or chlorobenzene, hydrocarbons such as benzene, xylene, toluene, hexane, cyclohexane or mineral oil fractions, or other solvents such as dimethylformamide (DMF), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU), N-methylpyrrolidone (NMP), acetonitrile or else water. It is equally possible to use mixtures of the solvents mentioned. Preference is given to dimethylformamide and toluene and also to a mixture of dimethylformamide and toluene.
Suitable bases for the process step (V)→(I-A) are alkali metal hydrides such as sodium hydride, alkali metal hydroxides such as, for example, lithium hydroxide, sodium hydroxide or potassium hydroxide, alkali metal carbonates such as lithium carbonate, sodium carbonate, potassium carbonate or caesium carbonate, alkali metal bicarbonates such as sodium bicarbonate or potassium bicarbonate, alkali metal alkoxides such as sodium methoxide or potassium methoxide, sodium ethoxide or potassium ethoxide or potassium tert-butoxide, or organic amines such as triethylamine, diisopropylethylamine, pyridine, 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU) or 1,5-diazabicyclo[4.3.0]non-5-ene (DBN). Preference is given to pyridine.
The reaction (V)→(I-A) is generally carried out in a temperature range of from −10° C. to +30° C., preferably from 0° C. to +20° C. The reaction can be carried out at atmospheric, elevated or reduced pressure (for example from 0.5 to 5 bar). The reaction is generally carried out at atmospheric pressure.
Inert solvents for process step (II)→(VI) are, for example, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, hydrocarbons such as benzene, xylene, toluene, hexane, cyclohexane or mineral oil fractions, or other solvents such as dimethylformamide (DMF), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU), N-methylpyrrolidone (NMP), pyridine or acetonitrile. It is equally possible to use mixtures of the solvents mentioned. Preference is given to DMSO.
The reaction (II)→(VI) is generally carried out in a temperature range of from +20° C. to +180° C., preferably from +100° C. to +160° C., if appropriate in a microwave. The reaction can be carried out at atmospheric, elevated or reduced pressure (for example from 0.5 to 5 bar). The reaction is generally carried out at atmospheric pressure.
The reaction (VI)→(VII) is carried out using methods known to the person skilled in the art in a two-step process initially with formation of the imino ester using sodium methoxide in methanol at from 0° C. to +40° C. and subsequent nucleophilic addition of an ammonia equivalent such as, for example, ammonia or ammonium chloride in acetic acid with formation of the amidine (VII) at from +50 to +150° C.
Inert solvents for process step (VII)+(VIII)→(IX) are alcohols such as methanol, ethanol, n-propanol, isopropanol, n-butanol or tert-butanol, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, hydrocarbons such as benzene, xylene, toluene, hexane, cyclohexane or mineral oil fractions, or other solvents such as dimethylformamide (DMF), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU), N-methylpyrrolidone (NMP), pyridine, acetonitrile or else water. It is equally possible to use mixtures of the solvents mentioned. Preference is given to DMF.
Suitable bases for the process step (VII)+(VIII)→(IX) are alkali metal hydroxides such as, for example, lithium hydroxide, sodium hydroxide or potassium hydroxide, alkali metal carbonates such as lithium carbonate, sodium carbonate, potassium carbonate or caesium carbonate, alkali metal bicarbonates such as sodium bicarbonate or potassium bicarbonate, alkali metal alkoxides such as sodium methoxide or potassium methoxide, sodium ethoxide or potassium ethoxide or potassium tert-butoxide, or organic amines such as triethylamine, diisopropylethylamine, pyridine, 1,8-diazabicyclo [5.4.0]undec-7-ene (DBU) or 1,5-diazabicyclo[4.3.0]non-5-ene (DBN). Preference is given to triethylamine.
The reaction (VII)+(VIII)→(IX) is generally carried out in a temperature range of from +20° C. to +150° C., preferably from +80° C. to +120° C., if appropriate in a microwave. The reaction can be carried out at atmospheric, elevated or reduced pressure (for example from 0.5 to 5 bar). The reaction is generally carried out at atmospheric pressure.
The compound of the formula (VIII) can be prepared analogously to the literature L. F. Cavalieri, J. F. Tanker, A. Bendich, J. Am. Chem. Soc., 1949, 71, 533.
Inert solvents for the reaction (I-A)→(I-B) are, for example, halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, trichloroethylene or chlorobenzene, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, or other solvents such as dimethylformamide (DMF), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU), N-methylpyrrolidone (NMP), pyridine or acetonitrile. Preference is given to tetrahydrofuran.
Suitable bases for the process step (I-A)→(I-B) are alkali metal hydrides such as potassium hydride or sodium hydride, alkali metal carbonates such as lithium carbonate, sodium carbonate, potassium carbonate or caesium carbonate, alkali metal bicarbonates such as sodium bicarbonate or potassium bicarbonate, alkali metal alkoxides such as sodium methoxide or potassium methoxide, sodium ethoxide or potassium ethoxide or potassium tert-butoxide, amides such as sodium amide, lithium bis(trimethylsilyl)amide, sodium bis(trimethylsilyl)amide or potassium bis(trimethylsilyl)amide or lithium diisopropylamide, organometallic compounds such as butyllithium or phenyllithium, or organic amines such as triethylamine, diisopropylethylamine, pyridine, 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU) or 1,5-diazabicyclo[4.3.0]non-5-ene (DBN). Preference is given to lithium bis(trimethylsilyl)amide, sodium bis(trimethylsilyl)amide and sodium hydride.
The reaction (I-A)→(I-B) is generally carried out in a temperature range of from −78° C. to +40° C., preferably from 0° C. to +20° C. The reaction can be carried out at atmospheric, elevated or reduced pressure (for example from 0.5 to 5 bar). The reaction is generally carried out at atmospheric pressure.
The preparation processes described can be illustrated in an exemplary manner by the synthesis schemes below (Schemes 1 to 3):
Figure US11439642-20220913-C00012
Figure US11439642-20220913-C00013
Figure US11439642-20220913-C00014
The compound of the formula (II) can be prepared by cyclizing the compound of the formula (X)
Figure US11439642-20220913-C00015

in an inert solvent with hydrazine hydrate to give the compound of the formula (XI)
Figure US11439642-20220913-C00016

then reacting this compound in an inert solvent in the presence of a suitable Lewis acid initially with isopentyl nitrite to give the corresponding diazonium salt and then converting this salt directly with sodium iodide into the compound of the formula (XII)
Figure US11439642-20220913-C00017

and then reacting this compound in an inert solvent in the presence of a suitable base with the compound of the formula (XIII)
Figure US11439642-20220913-C00018
Inert solvents for process step (X)→(XI) are alcohols such as methanol, ethanol, n-propanol, isopropanol, n-butanol, tert-butanol or 1,2-ethanediol, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, hydrocarbons such as benzene, xylene, toluene, hexane, cyclohexane or mineral oil fractions, or other solvents such as dimethylformamide (DMF), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU), N-methylpyrrolidone (NMP), pyridine, acetonitrile or else water. It is equally possible to use mixtures of the solvents mentioned. 1,2-Ethanediol is preferred.
The reaction (X)→(XI) is generally carried out in a temperature range of from +60° C. to +200° C., preferably from +120° C. to +180° C. The reaction can be carried out at atmospheric, elevated or reduced pressure (for example from 0.5 to 5 bar). The reaction is generally carried out at atmospheric pressure.
Inert solvents for the reaction (XI)→(XII) are, for example, halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, trichloroethylene or chlorobenzene, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, or other solvents such as dimethylformamide (DMF), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU), N-methylpyrrolidone (NMP), pyridine or acetonitrile. Preference is given to DMF.
Suitable Lewis acids for the process step (XI)→(XII) are boron trifluoride/diethyl ether complex, cerium(IV) ammonium nitrate (CAN), tin(II) chloride, lithium perchlorate, zinc(II) chloride, indium(III) chloride or indium(III) bromide. Preference is given to boron trifluoride/diethyl ether complex.
The reaction (XI)→(XII) is generally carried out in a temperature range of from −78° C. to +40° C., preferably from 0° C. to +20° C. The reaction can be carried out at atmospheric, elevated or reduced pressure (for example from 0.5 to 5 bar). The reaction is generally carried out at atmospheric pressure.
Inert solvents for the reaction (XII)+(XIII)→(II) are, for example, halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, trichloroethylene or chlorobenzene, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, or other solvents such as dimethylformamide (DMF), dimethyl sulphoxide (DMSO), N,N′-dimcthylpropylcncurca (DMPU), N-methylpyrrolidone (NMP), pyridine or acetonitrile. Preference is given to DMF.
Suitable bases for the process step (XII)+(XIII)→(II) are alkali metal hydrides such as potassium hydride or sodium hydride, alkali metal carbonates such as lithium carbonate, sodium carbonate, potassium carbonate or caesium carbonate, alkali metal bicarbonates such as sodium bicarbonate or potassium bicarbonate, alkali metal alkoxides such as sodium methoxide or potassium methoxide, sodium ethoxide or potassium ethoxide or potassium tert-butoxide, amides such as sodium amide, lithium bis(trimethylsilyl)amide, sodium bis(trimethylsilyl)amide or potassium bis(trimethylsilyl)amide or lithium diisopropylamide, organometallic compounds such as butyllithium or phenyllithium, or organic amines such as triethylamine, diisopropylethylamine, pyridine, 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU) or 1,5-diazabicyclo[4.3.0]non-5-ene (DBN). Preference is given to caesium carbonate.
The reaction (XII)+(XIII)→(II) is generally carried out in a temperature range of from 0° C. to +60° C., preferably from +10° C. to +25° C. The reaction can be carried out at atmospheric, elevated or reduced pressure (for example from 0.5 to 5 bar). The reaction is generally carried out at atmospheric pressure.
The preparation process described can be illustrated in an exemplary manner by the synthesis scheme below (Scheme 4):
Figure US11439642-20220913-C00019
The present invention furthermore provides the compound:
5-fluoro-1-(2-fluorobenzyl)-3-iodo-1H-pyrazolo[3,4-b]pyridine
Figure US11439642-20220913-C00020
The present invention furthermore provides the compound:
5-fluoro-3-iodo-1H-pyrazolo[3,4-b]pyridine
Figure US11439642-20220913-C00021
The compounds of the formulae (III) and (XIII) are commercially available, known from the literature or can be prepared in analogy to literature processes.
The compound of the formula (X) is known from the literature [cf., for example, Winn M., J. Med. Chem. 1993, 36, 2676-7688; EP 634 413-A1; CN 1613849-A; EP 1626045-A1; WO 2009/018415] and can be prepared in analogy to literature processes or as shown in the synthesis scheme below (Scheme 5):
Figure US11439642-20220913-C00022
The compounds according to the invention act as stimulators of soluble guanylate cyclase and have an identical or improved therapeutic profile compared to compounds known from the prior art, such as, for example, with respect to their in vivo properties such as, for example, their pharmacokinetic and pharmacodynamic behaviour and/or their dose-activity relationship and/or their safety profile. They are therefore suitable for the treatment and/or prophylaxis of diseases in man and animals.
The compounds according to the invention lead to vasorelaxation, to an inhibition of platelet aggregation and to a reduction in blood pressure, and also to an increase in coronary blood flow. These effects are mediated via direct stimulation of soluble guanylate cyclase and intracellular cGMP increase. Moreover, the compounds according to the invention enhance the effect of substances increasing the cGMP concentration, such as, for example, EDRF (endothelium-derived relaxing factor), NO donors, protoporphyrin IX, arachidonic acid or phenylhydrazine derivatives.
The compounds according to the invention are suitable for the treatment and/or prophylaxis of cardiovascular, pulmonary, thromboembolic and fibrotic disorders.
Accordingly, the compounds according to the invention can be used in medicaments for the treatment and/or prophylaxis of cardiovascular disorders such as, for example, hypertension, acute and chronic heart failure, coronary heart disease, stable and unstable angina pectoris, peripheral and cardiac vascular disorders, arrhythmias, atrial and ventricular arrhythmias and impaired conduction such as, for example, atrioventricular blocks degrees I-III (AB block supraventricular tachyarrhythmia, atrial fibrillation, atrial flutter, ventricular fibrillation, ventricular flutter, ventricular tachyarrhythmia, Torsade de pointes tachycardia, atrial and ventricular extrasystoles, AV-junctional extrasystoles, Sick-Sinus syndrome, syncopes, AV-nodal re-entry tachycardia, Wolff-Parkinson-White syndrome, of acute coronary syndrome (ACS), autoimmune cardiac disorders (pericarditis, endocarditis, valvolitis, aortitis, cardiomyopathies), shock such as cardiogenic shock, septic shock and anaphylactic shock, aneurysms, boxer cardiomyopathy (premature ventricular contraction (PVC)), for the treatment and/or prophylaxis of thromboembolic disorders and ischaemias such as myocardial ischaemia, myocardial infarction, stroke, cardiac hypertrophy, transient and ischaemic attacks, preeclampsia, inflammatory cardiovascular disorders, spasms of the coronary arteries and peripheral arteries, oedema formation such as, for example, pulmonary oedema, cerebral oedema, renal oedema or oedema caused by heart failure, peripheral circulatory disturbances, reperfusion damage, arterial and venous thromboses, microalbuminuria, myocardial insufficiency, endothelial dsfunction, to prevent restenoses, for example after thrombolysis therapies, percutaneous transluminal angioplasties (PTA), transluminal coronary angioplasties (PTCA), heart transplants and bypass operations, and also micro- and macrovascular damage (vasculitis), increased levels of fibrinogen and of low-density lipoprotein (LDL) and increased concentrations of plasminogen activator inhibitor 1 (PAI-1), and also for the treatment and/or prophylaxis of erectile dysfunction and female sexual dysfunction.
In the context of the present invention, the term heart failure also includes more specific or related types of disease, such as acute decompensated heart failure, right heart failure, left heart failure, global failure, ischaemic cardiomyopathy, dilated cardiomyopathy, hypertrophic cardiomyopathy, idiopathic cardiomyopathy, congenital heart defects, heart valve defects, heart failure associated with heart valve defects, mitral stenosis, mitral insufficiency, aortic stenosis, aortic insufficiency, tricuspid stenosis, tricuspid insufficiency, pulmonary valve stenosis, pulmonary valve insufficiency, combined heart valve defects, myocardial inflammation (myocarditis), chronic myocarditis, acute myocarditis, viral myocarditis, diabetic heart failure, alcoholic cardiomyopathy, cardiac storage disorders, and diastolic and systolic heart failure.
In addition, the compounds according to the invention can also be used for the treatment and/or prophylaxis of arteriosclerosis, impaired lipid metabolism, hypolipoproteinaemias, dyslipidaemias, hypertriglyceridaemias, hyperlipidaemias, hypercholesterolaemias, abetalipoproteinaemias, sitosterolaemia, xanthomatosis, Tangier disease, adipositas, obesity and of combined hyperlipidaemias and metabolic syndrome.
The compounds according to the invention can additionally be used for the treatment and/or prophylaxis of primary and secondary Raynaud's phenomenon, of microcirculation impairments, claudication, peripheral and autonomic neuropathies, diabetic microangiopathies, diabetic retinopathy, diabetic ulcers on the extremities, gangrene, CREST syndrome, erythematosis, onychomycosis, rheumatic disorders and for promoting wound healing.
The compounds according to the invention are furthermore suitable for treating urological disorders such as, for example, benign prostate syndrome (BPS), benign prostate hyperplasia (BPH), benign prostate enlargement (BPE), bladder outlet obstruction (BOO), lower urinary tract syndromes (LUTS, including Feline Urological Syndrome (FUS)), disorders of the urogenital system including neurogenic over-active bladder (OAB) and (IC), incontinence (UI) such as, for example, mixed urinary incontinence, urge urinary incontinence, stress urinary incontinence or overflow urinary incontinence (MUI, UUI, SUI, OUI), pelvic pain, benign and malignant disorders of the organs of the male and female urogenital system.
The compounds according to the invention are furthermore suitable for the treatment and/or prophylaxis of kidney disorders, in particular of acute and chronic renal insufficiency and acute and chronic renal failure. In the context of the present invention, the term renal insufficiency comprises both acute and chronic manifestations thereof, as well as underlying or related kidney diseases such as renal hypoperfusion, intradialytic hypotension, obstructive uropathy, glomerulopathies, glomerulonephritis, acute glomerulonephritis, glomerulosclerosis, tubulointerstitial diseases, nephropathic diseases such as primary and congenital kidney disease, nephritis, immunological kidney diseases such as kidney graft rejection and immunocomplex-induced kidney diseases, nephropathy induced by toxic substances, nephropathy induced by contrast agents, diabetic and non-diabetic nephropathy, pyelonephritis, renal cysts, nephrosclerosis, hypertensive nephrosclerosis and nephrotic syndrome, which can be characterized diagnostically for example by abnormally reduced creatinine and/or water excretion, abnormally raised blood concentrations of urea, nitrogen, potassium and/or creatinine, altered activity of renal enzymes such as, for example, glutamyl synthetase, altered urine osmolarity or urine volume, increased microalbuminurea, macroalbuminurea, laesions on glomerulae and arterioles, tubular dilatation, hyperphosphataemia and/or need for dialysis. The present invention also comprises the use of the compounds according to the invention for the treatment and/or prophylaxis of sequelae of renal insufficiency, for example pulmonary oedema, heart failure, uraemia, anaemia, electrolyte disturbances (for example hypercalaemia, hyponatraemia) and disturbances in bone and carbohydrate metabolism.
Furthermore, the compounds according to the invention are also suitable for the treatment and/or prophylaxis of asthmatic disorders, pulmonary arterial hypertension (PAH) and other forms of pulmonary hypertension (PH) including left-heart disease, HIV, sickle cell anaemia, thromboembolisms (CTEPH), sarkoidosis, COPD or pulmonary fibrosis-associated pulmonary hypertension, chronic-obstructive pulmonary disease (COPD), acute respiratory distress syndrome (ARDS), acute lung injury (ALI), alpha-1-antitrypsin deficiency (AATD), pulmonary fibrosis, pulmonary emphysema (for example pulmonary emphysema induced by cigarette smoke) and cystic fibrosis (CF).
The compounds described in the present invention also represent active compounds for controlling central nervous system diseases characterized by disturbances of the NO/cGMP system. They are suitable in particular for improving perception, concentration, learning or memory after cognitive impairments like those occurring in particular in association with situations/diseases/syndromes such as mild cognitive impairment, age-associated learning and memory impairments, age-associated memory losses, vascular dementia, craniocerebral trauma, stroke, dementia occurring after strokes (post stroke dementia), post-traumatic craniocerebral trauma, general concentration impairments, concentration impairments in children with learning and memory problems, Alzheimer's disease, Lewy body dementia, dementia with degeneration of the frontal lobes including Pick's syndrome, Parkinson's disease, progressive nuclear palsy, dementia with corticobasal degeneration, amyolateral sclerosis (ALS), Huntington's disease, demyelination, multiple sclerosis, thalamic degeneration, Creutzfeld-Jacob dementia, HIV dementia, schizophrenia with dementia or Korsakoff's psychosis. They are also suitable for the treatment and/or prophylaxis of central nervous system disorders such as states of anxiety, tension and depression, CNS-related sexual dysfunctions and sleep disturbances, and for controlling pathological disturbances of the intake of food, stimulants and addictive substances.
The compounds according to the invention are furthermore also suitable for controlling cerebral blood flow and thus represent effective agents for controlling migraine. They are also suitable for the prophylaxis and control of sequelae of cerebral infarct (Apoplexia cerebri) such as stroke, cerebral ischaemias and skull-brain trauma. The compounds according to the invention can likewise be employed for controlling states of pain and tinnitus.
In addition, the compounds according to the invention have antiinflammatory action and can therefore be used as antiinflammatory agents for the treatment and/or prophylaxis of sepsis (SIRS), multiple organ failure (MODS, MOF), inflammatory disorders of the kidney, chronic intestinal inflammations (IBD, Crohn's disease, UC), pancreatitis, peritonitis, rheumatoid disorders, inflammatory skin diseases and inflammatory eye diseases.
Furthermore, the compounds according to the invention can also be used for the treatment and/or prophylaxis of autoimmune diseases.
The compounds according to the invention are furthermore suitable for the treatment and/or prophylaxis of fibrotic disorders of the internal organs such as, for example, the lung, the heart, the kidney, the bone marrow and in particular the liver, and also dermatological fibroses and fibrotic eye disorders. In the context of the present invention, the term fibrotic disorders includes in particular the following terms: hepatic fibrosis, cirrhosis of the liver, pulmonary fibrosis, endomyocardial fibrosis, nephropathy, glomerulonephritis, interstitial renal fibrosis, fibrotic damage resulting from diabetes, bone marrow fibrosis and similar fibrotic disorders, scleroderma, morphea, keloids, hypertrophic scarring (also following surgical procedures), naevi, diabetic retinopathy, proliferative vitreoretinopathy and disorders of the connective tissue (for example sarcoidosis).
The compounds according to the invention are furthermore suitable for controlling postoperative scarring, for example as a result of glaucoma operations.
The compounds according to the invention can also be used cosmetically for ageing and keratinized skin.
Moreover, the compounds according to the invention are suitable for the treatment and/or prophylaxis of hepatitis, neoplasms, osteoporosis, glaucoma and gastroparesis.
The present invention further provides for the use of the compounds according to the invention for the treatment and/or prophylaxis of disorders, in particular the disorders mentioned above.
The present invention further provides the use of the compounds according to the invention for the treatment and/or prophylaxis of heart failure, angina pectoris, hypertension, pulmonary hypertension, ischaemias, vascular disorders, kidney failure, thromboembolic disorders, fibrotic disorders and arteriosclerosis.
The present invention further provides the compounds according to the invention for use in a method for the treatment and/or prophylaxis of heart failure, angina pectoris, hypertension, pulmonary hypertension, ischaemias, vascular disorders, kidney failure, thromboembolic disorders, fibrotic disorders and arteriosclerosis.
The present invention further provides for the use of the compounds according to the invention for producing a medicament for treatment and/or prophylaxis of disorders, in particular the disorders mentioned above.
The present invention further provides for the use of the compounds according to the invention for producing a medicament for treatment and/or prophylaxis of heart failure, angina pectoris, hypertension, pulmonary hypertension, ischaemias, vascular disorders, kidney failure, thromboembolic disorders, fibrotic disorders and arteriosclerosis.
The present invention further provides a method for treatment and/or prophylaxis of disorders, in particular the disorders mentioned above, using an effective amount of at least one of the compounds according to the invention.
The present invention further provides a method for treatment and/or prophylaxis of heart failure, angina pectoris, hypertension, pulmonary hypertension, ischaemias, vascular disorders, kidney failure, thromboembolic disorders, fibrotic disorders and arteriosclerosis using an effective amount of at least one of the compounds according to the invention.
The compounds according to the invention can be employed alone or, if required, in combination with other active compounds. The present invention further provides medicaments comprising at least one of the compounds according to the invention and one or more further active compounds, especially for the treatment and/or prophylaxis of the aforementioned disorders. Preferred examples of suitable active compound combinations include:
    • organic nitrates and NO donors, for example sodium nitroprusside, nitroglycerin, isosorbide mononitrate, isosorbide dinitrate, molsidomine or SIN-1, and inhaled NO;
    • compounds which inhibit the breakdown of cyclic guanosine monophosphate (cGMP), such as, for example, inhibitors of phosphodiesterases (PDE) 1, 2 and/or 5, in particular PDE 5 inhibitors such as sildenafil, vardenafil and tadalafil;
    • agents having an antithrombotic effect, for example and with preference from the group of platelet aggregation inhibitors, of anticoagulants or of profibrinolytic substances;
    • active compounds which lower blood pressure, for example and preferably from the group of calcium antagonists, angiotensin AII antagonists, ACE inhibitors, endothelin antagonists, renin inhibitors, alpha-receptor blockers, beta-receptor blockers, mineralocorticoid receptor antagonists, and of diuretics; and/or
    • active compounds which alter lipid metabolism, for example and with preference from the group of thyroid receptor agonists, cholesterol synthesis inhibitors such as, by way of example and preferably, HMG-CoA reductase inhibitors or squalene synthesis inhibitors, of ACAT inhibitors, CETP inhibitors, MTP inhibitors, PPAR-alpha, PPAR-gamma and/or PPAR-delta agonists, cholesterol absorption inhibitors, lipase inhibitors, polymeric bile acid adsorbents, bile acid reabsorption inhibitors and lipoprotein(a) antagonists;
Agents having antithrombotic activity preferably mean compounds from the group of platelet aggregation inhibitors, of anticoagulants or of profibrinolytic substances.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a platelet aggregation inhibitor such as, by way of example and preferably, aspirin, clopidogrel, ticlopidin or dipyridamol.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a thrombin inhibitor such as, by way of example and preferably, ximelagatran, dabigatran, melagatran, bivalirudin or clexane.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a GPIIb/IIIa antagonist such as, by way of example and preferably, tirofiban or abciximab.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a factor Xa inhibitor such as, by way of example and preferably, rivaroxab an (BAY 59-7939), DU-176b, apixab an, otamixab an, fidexab an, razaxab an, fondaparinux, idraparinux, PMD-3112, YM-150, KFA-1982, EMD-503982, MCM-17, MLN-1021, DX 9065a, DPC 906, JTV 803, SSR-126512 or SSR-128428.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with heparin or a low molecular weight (LMW) heparin derivative.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a vitamin K antagonist such as, by way of example and preferably, coumarin.
Agents which lower blood pressure are preferably understood to mean compounds from the group of calcium antagonists, angiotensin AII antagonists, ACE inhibitors, endothelin antagonists, renin inhibitors, alpha-receptor blockers, beta-receptor blockers, mineralocorticoid receptor antagonists, and the diuretics.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a calcium antagonist such as, by way of example and preferably, nifedipine, amlodipine, verapamil or diltiazem.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an alpha-1 receptor blocker such as, by way of example and preferably, prazosin.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a beta receptor blocker such as, by way of example and preferably, propranolol, atenolol, timolol, pindolol, alprenolol, oxprenolol, penbutolol, bupranolol, metipranolol, nadolol, mepindolol, carazalol, sotalol, metoprolol, betaxolol, celiprolol, bisoprolol, carteolol, esmolol, labetalol, carvedilol, adaprolol, landiolol, nebivolol, epanolol or bucindolol.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an angiotensin AII antagonist such as, by way of example and preferably, losartan, candesartan, valsartan, telmisartan or embusartan.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an ACE inhibitor such as, by way of example and preferably, enalapril, captopril, lisinopril, ramipril, delapril, fosinopril, quinopril, perindopril or trandopril.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an endothelin antagonist such as, by way of example and preferably, bosentan, darusentan, ambrisentan or sitaxsentan.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a renin inhibitor such as, for example and preferably, aliskiren, SPP-600 or SPP-800.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a mineralocorticoid receptor antagonist such as, for example and preferably, spironolactone or eplerenone.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a loop diuretic such as, for example, furosemide, torasemide, bumetanide and piretanide, with potassium-sparing diuretics such as, for example, amiloride and triamterene, with aldosterone antagonists such as, for example, spironolactone, potassium canrenoate and eplerenone and also thiazide diuretics such as, for example, hydrochlorothiazide, chlorthalidone, xipamide and indapamide.
Active compounds which alter lipid metabolism are preferably understood to mean compounds from the group of CETP inhibitors, thyroid receptor agonists, cholesterol synthesis inhibitors such as HMG-CoA reductase inhibitors or squalene synthesis inhibitors, of ACAT inhibitors, MTP inhibitors, PPAR-alpha, PPAR-gamma and/or PPAR-delta agonists, cholesterol absorption inhibitors, polymeric bile acid adsorbents, bile acid reabsorption inhibitors, lipase inhibitors and lipoprotein(a) antagonists;
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a CETP inhibitor such as, by way of example and preferably, dalcetrapib, BAY 60-5521, anacetrapib or CETP vaccine (CETi-1).
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a thyroid receptor agonist such as, by way of example and preferably, D-thyroxin, 3,5,3′-triiodothyronin (T3), CGS 23425 or axitirome (CGS 26214).
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a HMG-CoA reductase inhibitor from the class of the statins such as, by way of example and preferably, lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, rosuvastatin or pitavastatin.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a squalene synthesis inhibitor such as, by way of example and preferably, BMS-188494 or TAK-475.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an ACAT inhibitor such as, by way of example and preferably, avasimibe, melinamide, pactimibe, eflucimibe or SMP-797.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an MTP inhibitor such as, by way of example and preferably, implitapide, BMS-201038, R-103757 or JTT-130.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a PPAR-gamma agonist such as, by way of example and preferably, pioglitazonc or rosiglitazonc.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a PPAR-delta agonist such as, for example and preferably, GW 501516 or BAY 68-5042.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a cholesterol absorption inhibitor such as, by way of example and preferably, ezetimibe, tiqueside or pamaqueside.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a lipase inhibitor, a preferred example being orlistat.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a polymeric bile acid adsorbent such as, by way of example and preferably, cholestyramine, colestipol, colesolvam, CholestaGel or colestimide.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a bile acid reabsorption inhibitor such as, by way of example and preferably, ASBT (=IBAT) inhibitors, for example AZD-7806, S-8921, AK-105, BARI-1741, SC-435 or SC-635
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an lipoprotein(a) antagonist such as, by way of example and preferably, gemcabene calcium (CI-1027) or nicotinic acid.
The present invention further provides medicaments which comprise at least one compound according to the invention, typically together with one or more inert, nontoxic, pharmaceutically suitable auxiliaries, and the use thereof for the aforementioned purposes.
The compounds according to the invention may act systemically and/or locally. For this purpose, they can be administered in a suitable manner, for example by the oral, parenteral, pulmonal, nasal, sublingual, lingual, buccal, rectal, dermal, transdermal, conjunctival, otic route, or as an implant or stent.
The compounds according to the invention can be administered in administration forms suitable for these administration routes.
Suitable administration forms for oral administration are those which work according to the prior art, which release the compounds according to the invention rapidly and/or in a modified manner and which contain the compounds according to the invention in crystalline and/or amorphized and/or dissolved form, for example tablets (uncoated or coated tablets, for example with gastric juice-resistant or retarded-dissolution or insoluble coatings which control the release of the compound according to the invention), tablets or films/wafers which disintegrate rapidly in the oral cavity, films/lyophilizates or capsules (for example hard or soft gelatin capsules), sugar-coated tablets, granules, pellets, powders, emulsions, suspensions, aerosols or solutions.
Parenteral administration can bypass an absorption step (e.g. intravenously, intraarterially, intracardially, intraspinally or intralumbally) or include an absorption (e.g. intramuscularly, subcutaneously, intracutaneously, percutaneously or intraperitoneally). Administration forms suitable for parenteral administration include preparations for injection and infusion in the form of solutions, suspensions, emulsions, lyophilizates or sterile powders.
For the other administration routes, suitable examples are inhalable medicament forms (including powder inhalers, nebulizers), nasal drops, solutions or sprays, tablets, films/wafers or capsules for lingual, sublingual or buccal administration, suppositories, car or eye preparations, vaginal capsules, aqueous suspensions (lotions, shaking mixtures), lipophilic suspensions, ointments, creams, transdermal therapeutic systems (e.g. patches), milk, pastes, foams, sprinkling powders, implants or stents.
Oral or parenteral administration is preferred, especially oral administration.
The compounds according to the invention can be converted to the administration forms mentioned. This can be done in a manner known per se, by mixing with inert, nontoxic, pharmaceutically suitable excipients. These excipients include carriers (for example microcrystalline cellulose, lactose, mannitol), solvents (e.g. liquid polyethylene glycols), emulsifiers and dispersing or wetting agents (for example sodium dodecylsulphate, polyoxysorbitan oleate), binders (for example polyvinylpyrrolidone), synthetic and natural polymers (for example albumin), stabilizers (e.g. antioxidants, for example ascorbic acid), dyes (e.g. inorganic pigments, for example iron oxides) and flavour and/or odour correctants.
In general, it has been found to be advantageous in the case of parenteral administration to administer amounts of about 0.001 to 1 mg/kg, preferably about 0.01 to 0.5 mg/kg, of body weight to achieve effective results. In the case of oral administration, the dosage is about 0.01 to 100 mg/kg, preferably about 0.01 to 20 mg/kg and most preferably 0.1 to 10 mg/kg of body weight.
It may nevertheless be necessary where appropriate to deviate from the stated amounts, specifically as a function of the body weight, route of administration, individual response to the active compound, nature of the preparation and time or interval over which administration takes place. For instance, in some cases, less than the aforementioned minimum amount may be sufficient, while in other cases the upper limit mentioned must be exceeded. In the case of administration of relatively large amounts, it may be advisable to divide these into several individual doses over the course of the day.
The working examples which follow illustrate the invention. The invention is not limited to the examples.
The percentages in the tests and examples which follow are, unless indicated otherwise, percentages by weight; parts are parts by weight. Solvent ratios, dilution ratios and concentration data for liquid/liquid solutions are based in each case on volume.
A. EXAMPLES
Abbreviations and Acronyms:
  • aq. aqueous solution
  • calc. calculated
  • br s broad singlet (in NMR)
  • DCI direct chemical ionization (in MS)
  • dec. decomposition point
  • DMF dimethylformamide
  • DMSO dimethyl sulphoxide
  • DSC dynamic differential calorimetry
  • eq. equivalent(s)
  • ESI electrospray ionization (in MS)
  • Et ethyl
  • fnd. found
  • h hour(s)
  • HPLC high-pressure high-performance liquid chromatography
  • HRMS high-resolution mass spectrometry
  • conc. concentrated
  • LC-MS liquid chromatography-coupled mass spectrometry
  • LiHMDS lithium hexamethyldisilazide
  • Me methyl
  • min minute(s)
  • MS mass spectrometry
  • NMR nuclear magnetic resonance spectrometry
  • Pd2dba3 tris(dibenzylideneacetone)dipalladium
  • Ph phenyl
  • PLM polarized light microscope
  • RT room temperature
  • Rt retention time (in HPLC)
  • TGA thermogravimetric analysis
  • THF tetrahydrofuran
  • UV ultraviolet spectrometry
  • v/v volume to volume ratio (of a solution)
    LC/MS Methods:
Method 1: MS instrument type: Waters ZQ; apparatus type HPLC: Agilent 1100 Series; UV DAD; column: Thermo Hypersil GOLD 3μ 20 mm×4 mm; mobile phase A: 1 l of water+0.5 ml of 50% strength formic acid, mobile phase B: 1 l of acetonitrile+0.5 ml of 50% strength formic acid; gradient: 0.0 min 100% A 3.0 min 10% A 4.0 min 10% A 4.1 min 100% A (flow rate 2.5 ml/min); oven: 55° C.; flow rate: 2 ml/min; UV detection: 210 nm.
Method 2: Instrument: Waters ACQUITY SQD UPLC System; column: Waters Acquity UPLC HSS T3 1.8μ 50×1 mm; mobile phase A: 1 l of water+0.25 ml of 99% strength formic acid, mobile phase B: 1 l of acetonitrile+0.25 ml of 99% strength formic acid; gradient: 0.0 min 90% A→1.2 min 5% A→2.0 min 5% A oven: 50° C.; flow rate: 0.40 ml/min; UV detection: 210-400 nm.
Method 3: Instrument: Micromass Quattro Premier with Waters UPLC Acquity; column: Thermo Hypersil GOLD 1.9μ 50 mm×1 mm; mobile phase A: 1 l of water+0.5 ml of 50% strength formic acid, mobile phase B: 1 l of acetonitrile+0.5 ml of 50% strength formic acid; gradient: 0.0 min 90% A 0.1 min 90% A 1.5 min 10% A 2.2 min 10% A; oven 50° C.; flow rate: 0.33 ml/min;
UV detection: 210 nm.
General Methods:
PLM: The polarized light microscopy was carried out using a Clemex PS3 polarized light microscope particle size system with a Leica DM microscope procided with 50×, 100×, 200×, and 500× lenses, a high-resolution monochrome 1600×1200 pixel digital camera and a motorized X-Y Marzhauser station (controlled by a Clemex ST-2000 controller). The samples of the crystalline material were measured on a glass slide (76×26 mm) in a drop of oil, the sample being covered with a cover glass (22×40 mm).
DSC: The melting points were determined by dynamic differential calorimetry. The determination was carried out using a Mettler-Toledo 823e DSC instrument provided with a TSO801RO sample robot and STARe software. About 1.5 to 3 mg of the sample were weighed out into a small aluminium pan, which was then closed with a perforated cap. The heat flow was measured in a temperature range of from 30 to 400° C. at a heating rate of 10° C./min and under an argon stream of 30 ml/min.
TGA: The thermogravimetric analysis was carried out using a Mettler-Toledo TGA/SDTA851e TGA instrument provided with a TSO801RO sample robot and STARe software. About 1.5 to 3 mg of the sample were weighed out into a small open aluminium pan (100 The sample weight was measured in a temperature range of from 30 to 400° C. at a heating rate of 10° C./min and under an argon stream of 30 ml/min.
The elemental analyses were carried out by Currenta GmbH & Co. using methods known to the person skilled in the art, in accordance with industry norm DIN-ISO 17025.
Starting Materials and Intermediates:
Example 1A 2,6-Dichloro-5-fluoronicotinamide
Figure US11439642-20220913-C00023
A suspension of 25 g (130.90 mmol) of 2,6-dichloro-5-fluoro-3-cyanopyridine in conc. sulphuric acid (125 ml) was stirred at 60-65° C. for 1 h. After cooling to RT, the contents of the flask were poured into ice-water and extracted three times with ethyl acetate (100 ml each time). The combined organic phases were washed with water (100 ml) and then with saturated aqueous sodium hydrogen carbonate solution (100 ml), dried and concentrated on a rotary evaporator. The material obtained was dried under high vacuum.
Yield: 24.5 g (90% of theory)
1H NMR (400 MHz, DMSO-d6): δ=7.95 (br s, 1H), 8.11 (br s, 1H), 8.24 (d, 1H).
Example 2A 2-Chloro-5-fluoronicotinamide
Figure US11439642-20220913-C00024
At RT, 44 g (210.58 mmol) of 2,6-dichloro-5-fluoronicotinamide were added to a suspension of 21.9 g (335.35 mmol) of zinc in methanol (207 ml). Acetic acid (18.5 ml) was then added, and the mixture was heated with stirring at reflux for 24 h. The contents of the flask were then decanted from the zinc, and ethyl acetate (414 ml) and saturated aqueous sodium hydrogen carbonate solution (414 ml) were added, followed by intense extractive stirring. Subsequently the reaction mixture was filtered with suction through kieselguhr and the filter product was washed three times with ethyl acetate (517 ml each time). The organic phase was separated off and the aqueous phase was washed with ethyl acetate (258 ml). The combined organic phases were washed once with saturated aqueous sodium hydrogen carbonate solution (414 ml), dried and concentrated under reduced pressure. Dichloromethane (388 ml) was added to the crystals obtained in this manner, and the mixture was stirred for 20 min. The mixture was once more filtered off with suction, washed with diethyl ether and sucked dry.
Yield: 20.2 g (53% of theory)
1H NMR (400 MHz, DMSO-d6): δ=7.87 (br s, 1H), 7.99 (dd, 1H), 8.10 (br s, 1H), 8.52 (d, 1H).
Example 3A 2-Chloro-5-fluoronicotinonitrile
Figure US11439642-20220913-C00025
81.2 ml (582.25 mmol) of triethylamine were added to a suspension of 46.2 g (264.66 mmol) of 2-chloro-5-fluoronicotinamide in dichloromethane (783 ml), and the mixture was cooled to 0° C. Then, with stirring, 41.12 ml (291.13 mmol) of trifluoroacetic anhydride were added slowly dropwise and the mixture was stirred at 0° C. for 1.5 h. The reaction solution was subsequently washed twice with saturated aqueous sodium bicarbonate solution (391 ml each time), dried and concentrated under reduced pressure.
Yield: 42.1 g (90% of theory)
1H NMR (400 MHz, DMSO-d6): δ=8.66 (dd, 1H), 8.82 (d, 1H).
Example 4A 5-Fluoro-1H-pyrazolo[3,4-b]pyridine-3-amine
Figure US11439642-20220913-C00026
A suspension of 38.5 g (245.93 mmol) of 2-chloro-5-fluoronicotinonitrile was initially charged in 1,2-ethanediol (380 ml), and hydrazine hydrate (119.6 ml, 2.459 mol) was then added. The mixture was heated under reflux with stirring for 4 h. The product precipitated on cooling. Water (380 ml) was added to the yellow crystals, and the mixture was subjected to extractive stirring at RT for 10 min. The suspension was then filtered with suction over a frit, and the filter product was washed with water (200 ml) and with −10° C. cold THF (200 ml). The residue was dried under high vacuum over phosphorus pentoxide.
Yield: 22.8 g (61% of theory)
1H NMR (400 MHz, DMSO-d6): δ=5.54 (s, 2H), 7.96 (dd, 1H), 8.38 (m, 1H), 12.07 (m, 1H).
Example 5A 5-Fluoro-3-iodo-1H-pyrazolo[3,4-b]pyridine
Figure US11439642-20220913-C00027
10 g (65.75 mmol) of 5-fluoro-1H-pyrazolo[3,4-b]pyridine-3-amine were initially charged in THF (329 ml), and the mixture was cooled to 0° C. 16.65 ml (131.46 mmol) of boron trifluoride diethyl ether complex were then added slowly. The reaction mixture was cooled further to −10° C. A solution of 10.01 g (85.45 mmol) of isopentyl nitrite in THF (24.39 ml) was then added slowly, and the mixture was stirred for a further 30 min. The mixture was diluted with cold diethyl ether (329 ml) and the resulting solid was isolated by filtration. A little at a time, the diazonium salt thus prepared was added to a cold (0° C.) solution of 12.81 g (85.45 mmol) of sodium iodide in acetone (329 ml), and the mixture was stirred at RT for 30 min. The reaction mixture was poured into ice-water (1.8 l) and extracted twice with ethyl acetate (487 ml each time). The collected organic phases were washed with saturated aqueous sodium chloride solution (244 ml), dried, filtered and concentrated. This gave 12.1 g (86% purity, 60% of theory) of the desired compound in the form of a brown solid. The crude product was reacted without further purification.
LC-MS (method 1): Rt=1.68 min; MS (ESIpos): m/z=264 (M+H)+.
Example 6A 5-Fluoro-1-(2-fluorobenzyl)-3-iodo-1H-pyrazolo[3,4-b]pyridine
Figure US11439642-20220913-C00028
141 g (462.11 mmol) of the compound from Example 5A were introduced into DMF (2538 ml), and 96.09 g (508.32 mmol) of 2-fluorobenzyl bromide and 165.62 g (508.32 mmol) of caesium carbonate were then added. The mixture was stirred at RT for two hours. The reaction mixture was then poured into saturated aqueous sodium chloride solution (13 670 ml) and extracted twice with ethyl acetate (5858 ml). The collected organic phases were washed with saturated aqueous sodium chloride solution (3905 ml), dried, filtered and concentrated. The residue was chromatographed on silica gel (mobile phase: petroleum ether/ethyl acetate 97:3) and the product fractions were concentrated. The resulting solid was dissolved in dichloromethane and washed once with saturated aqueous sodium thiosulphate solution (500 ml) and then with saturated aqueous sodium chloride solution (500 ml). The product was concentrated to dryness and the residue was suspended in diethyl ether, isolated by filtration with suction and dried under high vacuum. This gave 106.6 g (62% of theory) of the desired compound.
1H NMR (400 MHz, DMSO-d6): δ=5.73 (s, 2H), 7.13-7.26 (m, 3H), 7.33-7.41 (m, 1H), 7.94 (dd, 1H), 8.69-8.73 (m, 1H).
Example 7A 2-[5-Fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]-5-nitropyrimidine-4,6-diamine
Figure US11439642-20220913-C00029
Under argon, 860 mg (2.32 mmol) of the compound from Example 6A were introduced into 1,4-dioxane (86 ml), and the reaction mixture was flushed with argon for 10 min. Then 3.51 ml (6.95 mmol) of hexabutylditin and 483 mg (2.55 mmol) of 2-chloro-5-nitropyrimidine-4,6-diamine (prepared by the method of Helvetica Chimica Acta (1951), 34, 835-40) were added. Subsequently 860 mg (0.744 mmol) of tetrakis(triphenylphosphine)palladium(0) were added and the reaction mixture was heated at reflux overnight. The mixture was then cooled to RT, water was added and the mixture was extracted twice with ethyl acetate. The collected organic phases were dried over sodium sulphate, filtered and concentrated. The residue was subjected to extractive stirring in ethyl acetate, and the solid was isolated by filtration and dried under high vacuum. This gave 355 mg (62% purity, 24% of theory) of the desired compound. The crude product was reacted without further purification.
LC-MS (method 2): Rt=1.03 min
MS (ESIpos): m/z=399 (M+H)+
Example 8A 5-Fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridine-3-carbonitrile
Figure US11439642-20220913-C00030
A suspension of 16.03 g (43.19 mmol) of 5-fluoro-1-(2-fluorobenzyl)-3-iodo-1H-pyrazolo[3,4-b]pyridine (Example 6A) and 4.25 g (47.51 mmol) of copper cyanide was initially charged in DMSO (120 ml) and stirred at 150° C. for 2 h. After cooling, the contents of the flask were cooled to about 40° C. and poured into a solution of conc. aqueous ammonia (90 ml) and water (500 ml), ethyl acetate (200 ml) was added and the mixture was subjected to brief extractive stirring. The aqueous phase was separated off and extracted two more times with ethyl acetate (200 ml each time). The combined organic phases were washed twice with 10% strength aqueous sodium chloride solution (100 ml each time), dried and concentrated under reduced pressure. The crude product was reacted without further purification.
Yield: 11.1 g (91% of theory)
1H NMR (400 MHz, DMSO-d6): δ=5.87 (s, 2H), 7.17-7.42 (m, 4H), 8.52 (dd, 1H), 8.87 (dd, 1H).
Example 9A 5-Fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridine-3-carboximidamide acetate
Figure US11439642-20220913-C00031
11.1 g (41.07 mmol) of 5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridine-3-carbonitrile (Example 8A) were added to 2.22 g (41.07 mmol) of sodium methoxide in methanol (270 ml), and the mixture was stirred at RT for 2 h. 2.64 g (49.29 mmol) of ammonium chloride and acetic acid (9.17 ml) were then added, and the mixture was heated at reflux overnight. The mixture was then concentrated to dryness and the residue was taken up in water (100 ml) and ethyl acetate (100 ml) and adjusted to a pH of 10 using 2N aqueous sodium hydroxide solution. The mixture was stirred intensively at RT for about 1 h. The resulting suspension was filtered with suction and the filter product was washed with ethyl acetate (100 ml), with water (100 ml) and once more with ethyl acetate (100 ml). The residue was dried under high vacuum over phosphorus pentoxide.
Yield: 9.6 g (78% of theory)
MS (ESIpos): m/z=288 (M+H)+
1H NMR (400 MHz, DMSO-d6): δ=1.85 (s, 3H), 5.80 (s, 2H), 7.14-7.25 (m, 3H), 7.36 (m, 1H), 8.42 (dd, 1H), 8.72 (dd, 1H).
Example 10A 2-[5-Fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]-5-[(E)-phenyldiazenyl]pyrimidine-4,6-diamine
Figure US11439642-20220913-C00032
With stirring, 3.85 g (41.34 mmol) of aniline were added to water (40 ml) and conc. hydrochloric acid (7.07 ml), and this mixture was cooled to 0° C. A solution of 2.85 g (41.34 mmol) of sodium nitrite in water (21 ml) was then added dropwise at between 0° C. and 5° C., followed by stirring at 0° C. for 15 min. Thereafter, at 0° C., a solution of 4.28 g (52.25 mmol) of sodium acetate in water (19 ml) was added rapidly dropwise, and then, with thorough stirring, a solution of 2.73 g (41.34 mmol) of malononitrile in ethanol (10 ml) was added dropwise. After 2 h at 0° C., the resulting precipitate was isolated by filtration with suction and washed three times with water (50 ml each time) and with petroleum ether (50 ml). The residue, still moist, was dissolved in DMF (46 ml) and added dropwise at precisely 85° C. to a solution of 9.5 g (33.07 mmol) of 5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridine-3-carboximidamide acetate (Example 9A) in DMF (46 ml) and triethylamine (5.76 ml). The mixture was then stirred at 100° C. for 4 h and left to cool to RT overnight. The mixture was poured into water (480 ml) and subjected to extractive stirring at RT for 1 h. After the precipitate had been isolated by filtration with suction, it was washed twice with water (100 ml each time) and twice with methanol (50 ml each time) and then dried under a high vacuum.
Yield: 9.6 g (59% of theory)
LC-MS (method 2): Rt=1.21 min
MS (ESIpos): m/z=458 (M+H)+
Example 11A 2-[5-Fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidine-4,5,6-triamine
Figure US11439642-20220913-C00033

Variant A: Preparation Starting from Example 7A:
In pyridine (30 ml), 378 mg (0.949 mmol) of the compound from Example 7A were introduced and then 143 mg (0.135 mmol) of palladium (10% on carbon) were added. The mixture was hydrogenated overnight at RT under standard hydrogen pressure. The suspension was then filtered through kieselguhr and the filtercake was washed with ethanol. The filtrate was concentrated and yielded 233 mg (81% purity, 51% of theory) of the desired compound, which was reacted without further purification.
Variant B: Preparation Starting from Example 10A:
In DMF (800 ml), 39.23 g (85.75 mmol) of the compound from Example 10A were introduced and then 4 g of palladium (10% on carbon) were added. The mixture was hydrogenated with stirring overnight under standard hydrogen pressure. The batch was filtered over kieselguhr and the filter product was washed with a little DMF and then with a little methanol, and concentrated to dryness. The residue was admixed with ethyl acetate and stirred vigorously, and the precipitate was filtered off with suction, washed with ethyl acetate and diisopropyl ether and dried under a high vacuum over Sicapent.
Yield: 31.7 g (100% of theory)
LC-MS (method 2): Rt=0.78 min
MS (ESIpos): m/z=369 (M+H)+
WORKING EXAMPLES Example 1 Methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate
Figure US11439642-20220913-C00034
In pyridine (600 ml), 31.75 g (86.20 mmol) of the compound from Example 11A were introduced under argon and cooled to 0° C. Then a solution of 6.66 ml (86.20 mmol) of methyl chloroformate in dichloromethane (10 ml) was added dropwise and the mixture was stirred at 0° C. for 1 h. Thereafter the reaction mixture was brought to RT, concentrated under reduced pressure and co-distilled repeatedly with toluene. The residue was stirred with water/ethanol and then filtered off on a frit, after which it was washed with ethanol and ethyl acetate. Subsequently the residue was again stirred with diethyl ether, isolated by filtration with suction and then dried under a high vacuum.
Yield: 24.24 g (65% of theory)
LC-MS (method 2): Rt=0.79 min
MS (ESIpos): m/z=427 (M+H)+
1H NMR (400 MHz, DMSO-d6): δ=3.62 (br. s, 3H), 5.79 (s, 2H), 6.22 (br. s, 4H), 7.10-7.19 (m, 2H), 7.19-7.26 (m, 1H), 7.32-7.40 (m, 1H), 7.67 and 7.99 (2 br. s, 1H), 8.66 (m, 1H), 8.89 (dd, 1H).
Example 2 Methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}methylcarbamate
Figure US11439642-20220913-C00035
A quantity of 200 mg (0.469 mmol) of methyl 4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-ylcarbamate (Example 1) was introduced in THF (5 ml) at 0° C. Then 0.704 ml (0.704 mmol) of lithium hexamethyldisilazane solution (1M in THF) was added and the mixture was stirred at this temperature for 20 min. Subsequently 43.8 μl (0.704 mmol) of iodomethane were added and the mixture was warmed to RT. After 1 h at this temperature, reaction was terminated with water (1 ml) and the reaction mixture was concentrated, the residue being separated by means of preparative RP-HPLC (water (+0.05% formic acid)-acetonitrile gradient).
Yield: 90 mg (44% of theory)
LC-MS (method 2): Rt=0.85 min
MS (ESIpos): m/z=441 (M+H)+
1H NMR (400 MHz, DMSO-d6): δ=3.00 (s, 3H), 3.53 and 3.66 (2 s, 3H), 5.81 (s, 2H), 6.57 (br. s, 4H), 7.13 (m, 2H), 7.22 (m, 1H), 7.35 (m, 1H), 8.67 (m, 1H), 8.87 (dd, 1H).
Example 3 Methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}(2,2,2-trifluoroethyl)carbamate
Figure US11439642-20220913-C00036
A quantity of 3.470 g (8.138 mmol) of the compound from Example 1 was suspended in 35 ml of THF, admixed at 0° C. with 358 mg (8.952 mmol) of sodium hydride (60% suspension in mineral oil) and stirred at 0° C. for 90 min, in the course of which a solution was formed. A quantity of 2.519 g (8.952 mmol) of 2,2,2-trifluoroethyl trichloromethanesulphonate was added and the mixture was stirred at RT for 48 h. It was then stirred with water and concentrated on a rotary evaporator. The residue was taken up in ethyl acetate, and the organic phase was washed twice with water and dried over sodium sulphate. This gave 5.005 g of the target compound (79% of theory, purity by HPLC 65%). A quantity of 250 mg of the residue was purified by means of preparative HPLC (mobile phase: methanol/water, gradient 30:70→90:10).
LC-MS (method 2): Rt=0.97 min; MS (EIos): m/z=509 (M+H)+.
1H NMR (400 MHz, DMSO-d6): δ [ppm]=3.63 (s, 3H), 4.06-4.15 (m, 2H), 5.80 (s, 2H), 6.46 (br s, 4H) 7.11-7.15 (m, 2H), 7.20-7.25 (m, 1H), 7.33-7.38 (m, 1H), 8.66 (dd, 1H), 8.91 (dd, 1H).
Example 4 Methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate hydrochloride
Figure US11439642-20220913-C00037
A solution of 100 mg (0.235 mmol) of Example 1 in 2 ml of 1,4-dioxane was prepared in a brown 5 ml glass bottle. In succession, 2 ml of isopropanol and 235 μl (0.235 mmol) of 1M hydrochloric acid were added to this solution, and the solution was stirred at RT until the solvents had evaporated. Air-drying gave 102 mg (94% of theory) of the title compound.
PLM (100×): crystalline
DSC: 224° C. (dec., ΔH=189 J/g)
TGA: 1% weight loss at 80° C.
LC-MS (method 3): Rt=0.91 min
MS (ESIpos): m/z=427 (M+H)+
1H NMR (400 MHz, DMSO-d6): δ [ppm]=3.35 and 3.65 (2 s, 3H), 5.92 (s, 2H), 7.15 (dd, 1H), 7.25 (m, 2H), 7.37 (m, 1H), 7.75 (br s, 4H), 8.08 and 8.39 (2 s, 1H), 8.82 (m, 2H), 13.2 (br s, 1H).
Elemental analysis for C19F16F2N8O2+HCl:
calculated: % C 49.31; % H 3.70; % N 24.21;
measured: % C 49.5; % H 3.7; % N 24.3.
Example 5 Methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate sulphate
Figure US11439642-20220913-C00038
A solution of 100 mg (0.235 mmol) of Example 1 in 2 ml of 1,4-dioxane was prepared in a brown 5 ml glass bottle. In succession, 2 ml of isopropanol and a solution of 938 μl (0.235 mmol) of 0.25M sulphuric acid were added to this solution, and the solution was stirred at RT until the solvents had evaporated. Air-drying gave 103 mg (83.7% of theory) of the title compound.
PLM (100×): crystalline
DSC: 242° C. (dec., ΔH=115 J/g)
TGA: no weight loss prior to decomposition
LC-MS (method 3): Rt=0.91 min
MS (ESIpos): m/z=427 (M+H)+
1H NMR (400 MHz, DMSO-d6): δ [ppm]=3.56 and 3.66 (2 s, 3H), 5.93 (s, 2H), 7.16 (m, 2H), 7.25 (dd, 1H), 7.38 (m, 1H), 7.59 (br s, 4H), 8.03 and 8.32 (2 s, 1H), 8.82 (m, 2H), 13.0 (br s, 1H).
Elemental analysis for C19H16F2N8O2+H2SO4:
calculated: % C 43.51; % H 3.46; % N 21.37;
measured: % C 43.6; % H 3.4; % N 21.2.
Example 6 Methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate phosphate
Figure US11439642-20220913-C00039
A solution of 100 mg (0.235 mmol) of Example 1 in 2 ml of 1,4-dioxane was prepared in a brown 5 ml glass bottle. In succession, 2 ml of THF and a solution of 16 μl (0.235 mmol) of 85% strength phosphoric acid in 0.3 ml of water were added to this solution, and the solution was stirred at RT until the solvents had evaporated. Air-drying gave 105 mg (85.4% of theory) of the title compound.
PLM (100×): crystalline
DSC: 183° C. (dec., ΔH=65 J/g)
TGA: 6% weight loss prior to decomposition
LC-MS (method 3): Rt=0.91 min
MS (ESIpos): m/z=427 (M+H)30
1H NMR (400 MHz, DMSO-d6): δ [ppm]=3.57 and 3.62 (2 s, methyl signal obscured by water signal, 3H), 5.79 (s, 2H), 6.22 (br s, 4H), 7.15 (m, 2H), 7.22 (dd, 1H), 7.36 (m, 1H), 7.67 and 7.99 (2 s, 1H), 8.66 (m, 1H), 8.90 (m, 1H).
31P-NMR (400 MHz, DMSO-d6): δ [ppm]=−1.1
Elemental analysis for C19F16F2N8O2+H3PO4+2 H2O:
calculated: % C 40.72; % H 4.14; % N 19.99;
measured: % C 40.5; % H 4.0; % N 19.5.
Example 7 Methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate mesylate
Figure US11439642-20220913-C00040
A solution of 100 mg (0.235 mmol) of Example 1 in 2 ml of 1,4-dioxane was prepared in a brown 5 ml glass bottle. In succession, 2 ml of ethanol and a solution of 22.5 mg (0.235 mmol) of methanesulphonic acid in 0.3 ml of water were added to this solution, and the solution was stirred at RT until the solvents had evaporated. Air-drying gave 103 mg (84% of theory) of the title compound.
PLM (100×): crystalline
DSC: 154° C. (ΔH=11.7 J/g), 167° C. (ΔH=−5 J/g), 215.2° C. (dec, ΔH=56.1 J/g)
TGA: gradual weight loss during the measurement
LC-MS (method 3): Rt=0.91 min
MS (ESIpos): m/z=427 (M+H)+
1H NMR (400 MHz, DMSO-d6): δ [ppm]=2.31 (s, 3H), 3.57 and 3.66 (2 s, 3H), 5.93 (s, 2H), 7.17 (m, 2H), 7.25 (dd, 1H), 7.39 (m, 1H), 7.66 (s br, 4H), 8.06 and 8.34 (2 s, 1H), 8.81 (dd, 1H), 8.83 (s, 1H), 13.0 (br s, 1H).
Elemental analysis for C19H16F2N8O2+CH4O3S+H2O:
calculated: % C 44.44; % H 4.14; % N 20.7;
measured: % C 44.3; % H 4.1; % N 20.2.
Example 8 Methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbonate ethane-1,2-disulphonate
Figure US11439642-20220913-C00041
A solution of 100 mg (0.235 mmol) of Example 1 in 2 ml of 1,4-dioxane was prepared in a brown 5 ml glass bottle. In succession, 2 ml of isopropanol and 44.6 mg (0.235 mmol) of ethane-1,2-disulphonic acid were added to this solution, and the solution was stirred at RT until the solvents had evaporated. Air-drying gave 111 mg (73.7% of theory) of the title compound.
PLM (100×): predominantly crystalline
DSC: 97° C. (dec., ΔH=103 J/g)
TGA: gradual weight loss during the measurement
LC-MS (method 3): Rt=0.90 min
MS (ESIpos): m/z=427 (M+H)+
1H NMR (400 MHz, DMSO-d6): δ [ppm]=2.66 (s, 4H), 3.57 and 3.66 (2 s, methyl signal obscured by water signal, 3H), 5.93 (s, 2H), 7.17 (m, 2H), 7.25 (m, 1H), 7.39 (m, 1H), 8.05 and 8.35 (2 s, 1H), 8.80 (dd, 1H), 8.84 (s, 1H).
Elemental analysis for C19H16F2N8O2+C2H6O6S2+0.25 H2O+0.25 C4HgO2:
calculated: % C 41.09; % H 3.84; % N 17.42;
measured: % C 41.2; % H 4.2; % N 17.6.
Example 9 Methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate maleate
Figure US11439642-20220913-C00042
A solution of 100 mg (0.235 mmol) of Example 1 in 2 ml of 1,4-dioxane was prepared in a brown 5 ml glass bottle. In succession, 2 ml of isopropanol and 27.2 mg (0.235 mmol) of maleic acid were added to this solution, and the solution was stirred at RT until the solvents had evaporated. Air-drying gave 108 mg (84.9% of theory) of the title compound.
PLM (100×): crystalline
DSC: 192° C. (dec., ΔH=173 J/g)
TGA: 3% weight loss prior to decomposition
LC-MS (method 3): Rt=0.91 min
MS (ESIpos): m/z=427 (M+H)+
1H NMR (400 MHz, DMSO-d6): δ [ppm]=3.56 and 3.64 (2 s, obscured by dioxane signal, 3H), 5.85 (s, 2H), 6.16 (s, 2H), 6.9 (br s, 4H), 7.15 (m, 2H), 7.23 (dd, 1H), 7.37 (m, 1H), 7.85 and 8.13 (2 s, 1H), 8.73 (s, 1H), 8.86 (dd, 1H).
Elemental analysis for C19H16F2N8O2+C4H4O4+0.5 H2O+0.5 C4H8O2:
calculated: % C 50.42; % H 4.23; % N 18.82;
measured: % C 50.7; % H 3.9; % N 18.8.
Example 10 Methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate nitrate
Figure US11439642-20220913-C00043
A solution of 100 mg (0.235 mmol) of Example 1 in 2 ml of isopropanol was prepared in a brown 5 ml glass bottle. In succession, 2 ml of isopropanol and 0.235 μl (0.235 mmol) of 1M nitric acid were added to this solution, and the solution was stirred at RT until the solvents had evaporated. Air-drying gave 103 mg (89.7% of theory) of the title compound.
PLM (100×): crystalline
DSC: 175° C. (dec., ΔH=−224 J/g)
TGA: 3% weight loss prior to decomposition
LC-MS (method 3): Rt=0.91 min
MS (ESIpos): m/z=427 (M+H)+
1H NMR (400 MHz, DMSO-d6): δ [ppm]=3.57 and 3.66 (2 s, 3H), 5.93 (s, 2H), 7.16 (m, 2H), 7.25 (dd, 1H), 7.38 (m, 1H), 7.65 (br s, 4H), 8.02 and 8.32 (2 s, 1H), 8.80 (dd, 1H), 8.83 (s, 1H), 13.0 (br s, 1H).
Elemental analysis for C19H16F2N8O2+HNO3+0.75 H2O:
calculated: % C 45.38; % H 3.71; % N 25.07;
measured: % C 45.4; % H 3.7; % N 25.0.
B. Assessment of Pharmacological Activity
The pharmacological effect of the compounds according to the invention can be shown in the following assays:
B-1. Vasorelaxant Effect In Vitro
Rabbits are stunned by a blow to the neck and exsanguinated. The aorta is removed, freed from adhering tissue and divided into rings of a width of 1.5 mm. The rings are placed individually under an initial tension in 5 ml organ baths with Krebs-Henseleit solution which is at 37° C., is gassed with carbogen and has the following composition (in each case mM): sodium chloride 119; potassium chloride: 4.8; calcium chloride dihydrate: 1; magnesium sulphate heptahydrate: 1.4; potassium dihydrogenphosphate: 1.2; sodium bicarbonate: 25; glucose: 10. The force of contraction is detected with Statham UC2 cells, amplified and digitized via A/D converters (DAS-1802 HC, Keithley Instruments, Munich) and recorded in parallel on chart recorders. To produce a contraction, phenylephrine is added to the bath cumulatively in increasing concentration. After several control cycles, the substance to be investigated is added in each further run in increasing dosage in each case, and the height of the contraction achieved is compared with the height of the contraction reached in the last preceding run. The concentration necessary to reduce the height of the control value by 50% is calculated from this (IC50 value). The standard administration volume is 5 μl and the proportion of DMSO in the bath solution corresponds to 0.1%.
Representative IC50 values for the compounds according to the invention are shown in the table below (Table 1):
TABLE 1
Example No. IC50 [nM]
1 958
2 251
3 515

B-2. Effect on a Recombinant Guanylate Cyclase Reporter Cell Line
The cellular activity of the compounds according to the invention is determined using a recombinant guanylate cyclase reporter cell line, as described in F. Wunder et al., Anal. Biochem. 339, 104-112 (2005).
Representative values (MEC=minimum effective concentration) for the compounds according to the invention are shown in the table below (Table 2):
TABLE 2
Example No. MEC [μM]
1 0.3
2 0.1
3 0.03

B-3. Radiotelemetric Measurement of Blood Pressure on Conscious Spontaneously Hypertensive Rats
A commercially available telemetry system from DATA SCIENCES INTERNATIONAL DSI, USA, is employed for the blood pressure measurements on conscious rats described below.
The system consists of 3 main components:
    • implantable transmitters (Physiotel® telemetry transmitter)
    • receivers (Physiotel® receiver) which are linked via a multiplexer (DSI Data Exchange Matrix) to a
    • data acquisition computer.
The telemetry system makes it possible to continuously record blood pressure, heart rate and body motions of conscious animals in their usual habitat.
Animal Material
The investigations are carried out on adult female spontaneously hypertensive rats (SHR Okamoto) with a body weight of >200 g. SHR/NCrl from the Okamoto Kyoto School of Medicine, 1963 were a cross of male Wistar Kyoto rats with highly elevated blood pressure and female rats having a slightly elevated blood pressure and at F13 handed over to the U.S. National Institutes of Health.
After transmitter implantation, the experimental animals are housed singly in type 3 Makrolon cages. They have free access to standard feed and water.
The day/night rhythm in the experimental laboratory is changed by the room lighting at 6.00 am and at 7.00 pm.
Transmitter Implantation
The telemetry transmitters TA11 PA-C40 used are surgically implanted under aseptic conditions in the experimental animals at least 14 days before the first experimental use. The animals instrumented in this way can be employed repeatedly after the wound has healed and the implant has settled.
For the implantation, the fasted animals are anaesthetized with pentobarbital (Nembutal, Sanofi: 50 mg/kg i.p.) and shaved and disinfected over a large area of their abdomens. After the abdominal cavity has been opened along the linea alba, the liquid-filled measuring catheter of the system is inserted into the descending aorta in the cranial direction above the bifurcation and fixed with tissue glue (VetBonD™, 3M). The transmitter housing is fixed intraperitoneally to the abdominal wall muscle, and layered closure of the wound is performed.
An antibiotic (Tardomyocel COMP, Bayer, 1 ml/kg s.c.) is administered postoperatively for prophylaxis of infection.
Substances and Solutions
Unless indicated otherwise, the substances to be investigated are administered orally by gavage in each case to a group of animals (n=6). The test substances are dissolved in suitable solvent mixtures, or suspended in 0.5% strength Tylose, appropriate for an administration volume of 5 ml/kg of body weight.
A solvent-treated group of animals is employed as control.
Test Procedure
The telemetry measuring unit present is configured for 24 animals. Each experiment is recorded under an experiment number (Vyear month day).
Each of the instrumented rats living in the system is assigned a separate receiving antenna (1010 Receiver, DSI).
The implanted transmitters can be activated externally by means of an incorporated magnetic switch and are switched to transmission in the run-up to the experiment. The emitted signals can be detected online by a data acquisition system (Dataquest™ A.R.T. for Windows, DSI) and be appropriately processed. The data are stored in each case in a file created for this purpose and bearing the experiment number.
In the standard procedure, the following are measured for 10-second periods in each case:
    • systolic blood pressure (SBP)
    • diastolic blood pressure (DBP)
    • mean arterial pressure (MAP)
    • heart rate (HR)
    • activity (ACT).
The acquisition of measured values is repeated under computer control at 5-minute intervals. The source data obtained as absolute value are corrected in the diagram with the currently measured barometric pressure (Ambient Pressure Reference Monitor; APR-1) and stored as individual data. Further technical details are given in the extensive documentation from the manufacturing company (DSI).
Unless indicated otherwise, the test substances are administered at 9.00 am on the day of the experiment. Following the administration, the parameters described above are measured over 24 hours.
Evaluation
After the end of the experiment, the acquired individual data are sorted using the analysis software (Dataquest™ A.R.T.™ Analysis). The blank value is assumed to be the time 2 hours before administration of the substance, so that the selected data set includes the period from 7.00 am on the day of the experiment to 9.00 am on the following day.
The data are smoothed over a presettable time by determination of the average (15-minute average) and transferred as a text file to a storage medium. The measured values presorted and compressed in this way are transferred into Excel templates and tabulated. For each day of the experiment, the data obtained are stored in a dedicated file carrying the number of the experiment. Results and test protocols are filed in paper form sorted by numbers.
Representative values for the compounds according to the invention are shown in the table below (Table 3):
TABLE 3
Example 1: Example 2:
Dosage Dosage Dosage
0.3 mg/kg 3.0 mg/kg 0.3 mg/kg
Vehicle p.o. p.o. Vehicle p.o.
hours mean mean mean hours mean mean
after blood blood blood after blood blood
substance pressure pressure pressure substance pressure pressure
administration (mm Hg) (mm Hg) (mm Hg) administration (mm Hg) (mm Hg)
0 153.6 151.0 149.0 0 149.0 161.3
1 164.5 148.4 129.3 1 158.2 145.7
2 146.7 136.4 111.1 2 142.2 130.5
3 145.4 130.6 106.0 3 149.2 121.5
4 149.6 129.1 107.8 4 152.3 123.1
5 149.9 132.8 109.3 5 155.8 121.6
6 151.6 125.6 106.8 6 147.3 123.8
7 147.6 131.9 110.9 7 147.3 124.4
8 147.5 131.8 109.8 8 149.3 128.7
9 150.8 138.5 114.3 9 151.0 133.7
10 149.8 138.3 114.5 10 152.5 139.2
11 154.0 138.9 115.6 11 150.3 137.9
12 145.3 137.7 118.8 12 146.2 143.0
13 141.1 142.9 120.4 13 143.2 146.0
14 147.8 144.5 122.8 14 146.4 149.2
15 151.0 143.8 125.8 15 150.5 152.3
16 151.3 146.3 131.5 16 145.3 155.5
17 148.8 141.8 124.7 17 143.9 156.3
18 149.2 138.4 129.6 18 150.3 157.3
19 151.2 149.2 135.6 19 147.7 156.9
20 152.6 145.1 135.2 20 153.4 156.3
21 146.3 142.1 129.3 21 148.6 149.3
22 146.3 141.8 128.3 22 153.3 147.1
23 150.3 143.6 130.2 23 151.1 153.1
24 147.4 135.1 130.8 24 154.1 152.3
REFERENCES
  • Klaus Witte, Kai Hu, Johanna Swiatek, Claudia Müssig, Georg Ertl and Björun Lemmer: Experimental heart failure in rats: effects on cardiovascular circadian rhythms and on myocardial β-adrenergic signaling. Cardiovasc Res 47 (2): 203-405, 2000; Kozo Okamoto: Spontaneous hypertension in rats. Int Rev Exp Pathol 7: 227-270, 1969; Maarten van den Buuse: Circadian Rhythms of Blood Pressure, Heart Rate, and Locomotor Activity in Spontaneously Hypertensive Rats as Measured With Radio-Telemetry. Physiology & Behavior 55(4): 783-787, 1994
    B-4. Determination of Pharmacokinetic Parameters Following Intravenous and Peroral Administration:
The pharmacokinetic parameters of the substance are determined in male CD-1 mice, male Wistar rats and female beagles. The administration volume is 10 ml/kg for mice, 5 ml/kg for rats and 0.5 ml/kg for dogs. Intravenous administration is via a formulation of species-specific plasma/DMSO (99/1) in the case of mice and rats and via water/PEG400/ethanol (50/40/10) in the case of dogs. For easier removal of blood, a silicone catheter is inserted into the right Vena jugularis externa of the rats before the administration of substance. The surgical intervention takes place one day prior to the experiment with isofluran anaesthesia and administration of an analgetic (atropine/rimadyl (3/1) 0.1 ml s.c.). Substance administration is as i.v. bolus in the case of mice, as i.v. bolus or via a 15-minute infusion in the case of rats and via a 15-minute infusion in the case of dogs. Removal of blood is after 0.033, 0.083, 0.17, 0.5, 1, 2, 3, 4, 6, 7 and 24 hours in the case of mice and, after a 15-minute infusion, after 0.083, 0.25, 0.28 0.33, 0.42, 0.75, 1, 2, 3, 4, 6, 7 and 24 hours in the case of dogs and rats and after an i.v. bolus administration, after 0.033, 0.083, 0.17, 0.5, 1, 2, 3, 4, 6, 7 and 24 hours in the case of rats. For all species, oral administration of the dissolved substance via gavage is carried out based on a water/PEG400/ethanol formulation (50/40/10). Here, the removal of blood from rats and dogs is after 0.083, 0.17, 0.5, 0.75, 1, 2, 3, 4, 6, 7 and 24 hours. The blood is removed into heparinized tubes. The blood plasma is then obtained by centrifugation; if required, it can be stored at −20° C. until further processing.
An internal standard (ZK 228859) is added to the unknown samples, calibration samples and QCs, and the protein is precipitated using excess acetonitrile. After addition of an ammonium acetate buffer (0.01 M, pH 6.8 (Example 1/3) or pH 3.0 (Example 2)) and subsequent vortexing, the mixture is centrifuged at 1000 g and the supernatant is examined by LC-MS/MS (API 4000, AB Sciex). Chromatographic separation is carried out on an Agilent 1100-HPLC. The injection volume is 10 μl. The separation column used is a Phenomenex Luna 5μ C8(2) 100 A 50×2 mm, adjusted to a temperature of 40° C. For Example 1, a binary mobile phase gradient at 400 μl/min is used (A: 0.01M ammonium acetate buffer pH 6.8, B: 0.1% formic acid in acetonitrile): 0 min (90% A), 1 min (90% A), 3.5 min (15% A), 4.5 min (15% A), 4.6 min (90% A), 7 min (90% A). For Example 2, a binary mobile phase gradient at 500 μl/min is used instead (A: 0.01M ammonium acetate buffer pH 3.0, B: 0.1% formic acid in acetonitrile): 0 min (90% A), 1.5 min (90% A), 3.5 min (10% A), 4.5 min (10% A), 5 min (90% A), 7 min (90% A). For Example 3, a binary mobile solvent gradient at 500 μl/min is used instead (A: 0.01M ammonium acetate buffer pH 6.8, B: 0.1% formic acid in acetonitrile): 0 min (90% A), 1 min (90% A), 3 min (10% A), 4 min (10% A), 4.5 min (90% A), 6 min (90% A). The temperature of the Turbo V ion source is 500° C. The following MS instrument parameters are used: curtain gas 20 units (Example 1), 16 units (Example 2) or 15 units (Example 3), ion spray voltage 5 kV (Example 1/2) or 4.5 units (Example 3), gas 1 35 units (Example 1/3) or 25 units (Example 2), gas 2 30 units, CAD gas 4 units (Example 1/3) or 3 units (Example 2). The substances are quantified by peak heights or areas using extracted ion chromatograms of specific MRM experiments.
The plasma concentration/time plots determined are used to calculate the pharmacokinetic parameters such as AUC, Cmax, MRT (mean residence time), t1/2 (half life) and CL (clearance) employing the validated pharmacokinetic calculation programs KinEx (Vers. 2.5 and 3).
As the substance quantification is carried out in plasma, it is necessary to determine the blood/plasma distribution of the substance to be able to adjust the pharamacokinetic parameters in an appropriate manner. To this end, a defined amount of substance is incubated in heparinized whole blood of the species in question in a rocking roller mixer for 20 min. After centrifugation at 1000 g, the plasma concentration is measured (see above) and determined by calculating the quotient of the cb/cp values.
Following intravenous administration of 0.3 mg/kg of the compounds according to the invention in rats, the following values were recorded:
Example
1.* 2.** 3.**
AUCnorm [kg · h/l] 4.36 1.79 1.36
CLblood [lh/kg] 0.29 0.53 1.02
MRT [h] 4.1 2.3 2.3
t1/2 [h] 3.4 1.7 1.9
*15-minute infusion
**i.v. bolus administration

B-5. Safety Profile
The substances according to the invention show a surprisingly favourable safety profile in vivo which was established by non-clinical safety studies according to OECD (OECD guidelines for testing of chemicals, No. 407) and ICH (3BS2A) guidelines.
C. Working Examples for Pharmaceutical Compositions
The compounds according to the invention can be converted to pharmaceutical formulations as follows
Tablet:
Composition:
100 mg of the compound according to the invention, 50 mg of lactose (monohydrate), 50 mg of maize starch (native), 10 mg of polyvinylpyrrolidone (PVP 25) (BASF, Ludwigshafen, Germany) and 2 mg of magnesium stearate.
Tablet weight 212 mg, diameter 8 mm, radius of curvature 12 mm.
Preparation:
The mixture of the compound according to the invention, lactose and starch is granulated with a 5% solution (w/w) of the PVP in water. After drying, the granules are mixed with the magnesium stearate for 5 minutes. This mixture is pressed with a conventional tableting press (for tablet dimensions see above). The guide value used for the pressing is a pressing force of 15 kN.
Suspension for Oral Administration:
Composition:
1000 mg of the compound according to the invention, 1000 mg of ethanol (96%), 400 mg of Rhodigcl® (xanthan gum from FMC, Pennsylvania, USA) and 99 g of water.
A single dose of 100 mg of the compound according to the invention corresponds to 10 ml of oral suspension.
Preparation:
The Rhodigel is suspended in ethanol and the compound according to the invention is added to the suspension. The water is added while stirring. The mixture is stirred for approx. 6 h until swelling of the Rhodigel has ended.
Solution for Oral Administration:
Composition:
500 mg of the compound according to the invention, 2.5 g of polysorbate and 97 g of polyethylene glycol 400. A single dose of 100 mg of the compound according to the invention corresponds to 20 g of oral solution.
Preparation:
The compound according to the invention is suspended in the mixture of polyethylene glycol and polysorbate while stirring. The stirring operation is continued until dissolution of the compound according to the invention is complete.
i.v. Solution:
The compound according to the invention is dissolved in a concentration below the saturation solubility in a physiologically acceptable solvent (e.g. isotonic saline, glucose solution 5% and/or PEG 400 solution 30%). The solution is subjected to sterile filtration and dispensed into sterile and pyrogen-free injection vessels.

Claims (66)

The invention claimed is:
1. A method of treating or preventing one or more of coronary heart disease, systolic or diastolic heart failure, angina pectoris, ischaemias, peripheral and cardiac vascular disorders, kidney failure, renal insufficiency, and increased microalbuminurea, comprising administering a therapeutically effective amount of a compound of formula (I-A)
Figure US11439642-20220913-C00044
or a salt thereof to a human or animal in need thereof.
2. The method of claim 1, comprising administering a therapeutically effective amount of the compound of formula (I-A)
Figure US11439642-20220913-C00045
3. The method of claim 1, comprising administering a therapeutically effective amount of the salt of the compound of formula (I-A)
Figure US11439642-20220913-C00046
4. The method of claim 1, wherein the method is for treating angina pectoris.
5. The method of claim 2, wherein the method is for treating angina pectoris.
6. The method of claim 3, wherein the method is for treating angina pectoris.
7. The method of claim 1, wherein the method is for treating vascular disorders.
8. The method of claim 2, wherein the method is for treating vascular disorders.
9. The method of claim 3, wherein the method is for treating vascular disorders.
10. The method of claim 1, wherein the method is for treating kidney failure.
11. The method of claim 2, wherein the method is for treating kidney failure.
12. The method of claim 3, wherein the method is for treating kidney failure.
13. The method of claim 1, wherein the method further comprises administering a further active compound that is a cGMP-PDE inhibitor selected from the group consisting of sildenafil, vardenafil and tadalafil.
14. The method of claim 2, wherein the method further comprises administering a further active compound that is a cGMP-PDE inhibitor selected from the group consisting of sildenafil, vardenafil and tadalafil.
15. The method of claim 3, wherein the method further comprises administering a further active compound that is a cGMP-PDE inhibitor selected from the group consisting of sildenafil, vardenafil and tadalafil.
16. The method of claim 1, wherein the method further comprises administering a further active compound that is an agent having blood pressure lowering activity selected from the group consisting of losartan, candesartan, valsartan, telmisartan, embusartan, bosentan, darusentan, ambrisentan, sitaxsentan, and spironolactone and eplerenone.
17. The method of claim 2, wherein the method further comprises administering a further active compound that is an agent having blood pressure lowering activity selected from the group consisting of losartan, candesartan, valsartan, telmisartan, embusartan, bosentan, darusentan, ambrisentan, sitaxsentan, and spironolactone and eplerenone.
18. The method of claim 3, wherein the method further comprises administering a further active compound that is an agent having blood pressure lowering activity selected from the group consisting of losartan, candesartan, valsartan, telmisartan, embusartan, bosentan, darusentan, ambrisentan, sitaxsentan, and spironolactone and eplerenone.
19. The method of claim 1, wherein the method is for treating coronary heart disease.
20. The method of claim 2, wherein the method is for treating coronary heart disease.
21. The method of claim 3, wherein the method is for treating coronary heart disease.
22. The method of claim 1, wherein the method is for treating systolic or diastolic heart failure.
23. The method of claim 2, wherein the method is for treating systolic or diastolic heart failure.
24. The method of claim 3, wherein the method is for treating systolic or diastolic heart failure.
25. The method of claim 1, wherein the method is for treating renal insufficiency.
26. The method of claim 2, wherein the method is for treating renal insufficiency.
27. The method of claim 3, wherein the method is for treating renal insufficiency.
28. The method of claim 1, wherein the method is for treating increased microalbuminurea.
29. The method of claim 2, wherein the method is for treating increased microalbuminurea.
30. The method of claim 3, wherein the method is for treating increased microalbuminurea.
31. A method of treatment of heart failure, hypertension, or pulmonary hypertension in humans and animals comprising administering an effective amount of a compound of formula (I-A)
Figure US11439642-20220913-C00047
or a salt thereof to a human or animal in need thereof.
32. The method of claim 31, wherein the method is for treatment of heart failure.
33. The method of claim 31, wherein the method is for treatment of hypertension.
34. The method of claim 31, wherein the method is for treatment of pulmonary hypertension.
35. The method of claim 31, wherein an effective amount of the salt of the compound of formula (I-A) is administered.
36. The method of claim 32, wherein an effective amount of the salt of the compound of formula (I-A) is administered.
37. The method of claim 33, wherein an effective amount of the salt of the compound of formula (I-A) is administered.
38. The method of claim 34, wherein an effective amount of the salt of the compound of formula (I-A) is administered.
39. A method of treating or preventing one or more of hypertension, pulmonary hypertension, heart failure, coronary heart disease, systolic or diastolic heart failure, angina pectoris, ischaemias, peripheral and cardiac vascular disorders, kidney failure, renal insufficiency, and increased microalbuminurea, comprising administering to a human or animal in need thereof therapeutically effective amounts of
(1) a compound of formula (I-A)
Figure US11439642-20220913-C00048
or a salt thereof; and
(2) a further active compound selected from the group consisting of organic nitrates, NO donors, cGMP-PDE inhibitors, agents having antithrombotic activity, agents lowering blood pressure, and agents altering lipid metabolism.
40. The method of claim 39, wherein an effective amount of the compound of formula (I-A) is administered.
41. The method of claim 39, wherein an effective amount of the salt of the compound of formula (I-A) is administered.
42. The method of claim 39, wherein the further active compound is selected from the group consisting of sodium nitroprusside, nitroglycerin, isosorbide mononitrate, isosorbide dinitrate, molsidomine or SIN-1, inhaled NO, inhibitors of phosphodiesterases (PDE) 1, 2 and/or 5, platelet aggregation inhibitors, anticoagulants, profibrinolytic substances, calcium antagonists, angiotensin AII antagonists, ACE inhibitors, endothelin antagonists, renin inhibitors, alpha-receptor blockers, beta-receptor blockers, mineralocorticoid receptor antagonists, diuretics, thyroid receptor agonists, HMG-CoA reductase inhibitors, squalene synthesis inhibitors, ACAT inhibitors, CETP inhibitors, MTP inhibitors; PPAR-alpha, PPAR-gamma and/or PPAR-delta agonists, cholesterol absorption inhibitors, lipase inhibitors, polymeric bile acid adsorbents, bile acid reabsorption inhibitors and lipoprotein(a) antagonists.
43. The method of claim 42, wherein the inhibitor of phosphodiesterases (PDE) 1, 2 and/or 5 is selected from the group consisting of sildenafil, vardenafil, and tadalafil.
44. The method of claim 42, wherein the angiotensin AII antagonist is selected from the group consisting of losartan, candesartan, valsartan, telmisartan, and embusartan.
45. The method of claim 42, wherein the endothelin antagonist is selected from the group consisting of bosentan, darusentan, ambrisentan, and sitaxsentan.
46. The method of claim 42, wherein the mineralocorticoid receptor antagonist is selected from the group consisting of spironolactone or eplerenone.
47. The method of claim 42, wherein an effective amount of the compound of formula (I-A) is administered.
48. The method of claim 42, wherein an effective amount of the salt of the compound of formula (I-A) is administered.
49. A method of treating or preventing one or more of vascular disorders, thromboembolic disorders, fibrotic disorders, and arteriosclerosis comprising administering a therapeutically effective amount of a compound of formula (I)
Figure US11439642-20220913-C00049
in which
R1 represents hydrogen or (C1-C4)-alkyl,
where (C1-C4)-alkyl may be substituted by one or two substituents independently of one another selected from the group consisting of fluorine and trifluoromethyl,
or an N-oxide, salt or salt of an N-oxide thereof to a human or animal in need thereof.
50. The method of claim 49, wherein in the compound of formula (I)
Rl represents hydrogen or methyl,
where methyl may be substituted by a trifluoromethyl substituent.
51. The method of claim 49, wherein the compound of formula (I) is selected from the group consisting of:
methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate
methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}1methylcarbamate
methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}(2,2,2-trifluoroethyl)carbamate
methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate hydrochloride
methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate sulphate
methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate phosphate
methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate mesylate
methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate ethane-1,2-disulphonate
methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate maleate and
methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate nitrate.
52. The method of claim 49, wherein the compound of formula (I) is:
Figure US11439642-20220913-C00050
or an N-oxide, salt, or salt of an N-oxide thereof.
53. The method of claim 49, wherein the method is for treating or preventing vascular disorders.
54. The method of claim 49, wherein the method is for treating or preventing thromboembolic disorders.
55. The method of claim 49, wherein the method is for treating or preventing fibrotic disorders.
56. The method of claim 49, wherein the method is for treating or preventing arteriosclerosis.
57. The method of claim 49, wherein the method further comprises administering a further active compound selected from the group consisting of organic nitrates, NO donors, cGMP-PDE inhibitors, agents having antithrombotic activity, agents having blood pressure lowering activity, and agents having lipid metabolism altering activity.
58. The method of claim 57, wherein the further active compound is an organic nitrate.
59. The method of claim 57, wherein the further active compound is an NO donor.
60. The method of claim 57, wherein the further active compound is a cGMP-PDE inhibitor.
61. The method of claim 57, wherein the further active compound is an agent having antithrombotic activity.
62. The method of claim 57, wherein the further active compound is an agent having blood pressure lowering activity.
63. The method of claim 57, wherein the further active compound is an agent having lipid metabolism altering activity.
64. The method of claim 49, wherein an effective amount of a compound of formula (I-A)
Figure US11439642-20220913-C00051
or a salt thereof is administered.
65. The method of claim 64, wherein an effective amount of the compound of formula (I-A) is administered.
66. The method of claim 64, wherein an effective amount of the salt of the compound of formula (I-A) is administered.
US16/919,385 2010-05-26 2020-07-02 Substituted 5-fluoro-1H-pyrazolopyridines and their use Active US11439642B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US16/919,385 US11439642B2 (en) 2010-05-26 2020-07-02 Substituted 5-fluoro-1H-pyrazolopyridines and their use
US17/820,667 US20220409618A1 (en) 2010-05-26 2022-08-18 Substituted 5-fluoro-1h-pyrazolopyridines and their use

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
DE102010021637A DE102010021637A1 (en) 2010-05-26 2010-05-26 Substituted 5-fluoro-1H-pyrazolopyridines and their use
DE102010021637.2 2010-05-26
US13/111,856 US8420656B2 (en) 2010-05-26 2011-05-19 Substituted 5-fluoro-1H-pyrazolopyridines and their use
US13/851,373 US8921377B2 (en) 2010-05-26 2013-03-27 Substituted 5-fluoro-1H-pyrazolopyridines and their use
US14/552,122 US9266885B2 (en) 2010-05-26 2014-11-24 Substituted 5-fluoro-1H-pyrazolopyridines and their use
US14/996,678 US20160129004A1 (en) 2010-05-26 2016-01-15 Substituted 5-fluoro-1h-pyrazolopyridines and their use
US15/348,545 US9993476B2 (en) 2010-05-26 2016-11-10 Substituted 5-flouro-1H-pyrazolopyridines and their use
US15/981,598 US10736896B2 (en) 2010-05-26 2018-05-16 Substituted 5-fluoro-1H-pyrazolopyridines and their use
US16/919,385 US11439642B2 (en) 2010-05-26 2020-07-02 Substituted 5-fluoro-1H-pyrazolopyridines and their use

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/981,598 Division US10736896B2 (en) 2010-05-26 2018-05-16 Substituted 5-fluoro-1H-pyrazolopyridines and their use

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/820,667 Continuation US20220409618A1 (en) 2010-05-26 2022-08-18 Substituted 5-fluoro-1h-pyrazolopyridines and their use

Publications (2)

Publication Number Publication Date
US20200397785A1 US20200397785A1 (en) 2020-12-24
US11439642B2 true US11439642B2 (en) 2022-09-13

Family

ID=44119218

Family Applications (8)

Application Number Title Priority Date Filing Date
US13/111,856 Active US8420656B2 (en) 2010-05-26 2011-05-19 Substituted 5-fluoro-1H-pyrazolopyridines and their use
US13/851,373 Active US8921377B2 (en) 2010-05-26 2013-03-27 Substituted 5-fluoro-1H-pyrazolopyridines and their use
US14/552,122 Active US9266885B2 (en) 2010-05-26 2014-11-24 Substituted 5-fluoro-1H-pyrazolopyridines and their use
US14/996,678 Abandoned US20160129004A1 (en) 2010-05-26 2016-01-15 Substituted 5-fluoro-1h-pyrazolopyridines and their use
US15/348,545 Active US9993476B2 (en) 2010-05-26 2016-11-10 Substituted 5-flouro-1H-pyrazolopyridines and their use
US15/981,598 Active US10736896B2 (en) 2010-05-26 2018-05-16 Substituted 5-fluoro-1H-pyrazolopyridines and their use
US16/919,385 Active US11439642B2 (en) 2010-05-26 2020-07-02 Substituted 5-fluoro-1H-pyrazolopyridines and their use
US17/820,667 Pending US20220409618A1 (en) 2010-05-26 2022-08-18 Substituted 5-fluoro-1h-pyrazolopyridines and their use

Family Applications Before (6)

Application Number Title Priority Date Filing Date
US13/111,856 Active US8420656B2 (en) 2010-05-26 2011-05-19 Substituted 5-fluoro-1H-pyrazolopyridines and their use
US13/851,373 Active US8921377B2 (en) 2010-05-26 2013-03-27 Substituted 5-fluoro-1H-pyrazolopyridines and their use
US14/552,122 Active US9266885B2 (en) 2010-05-26 2014-11-24 Substituted 5-fluoro-1H-pyrazolopyridines and their use
US14/996,678 Abandoned US20160129004A1 (en) 2010-05-26 2016-01-15 Substituted 5-fluoro-1h-pyrazolopyridines and their use
US15/348,545 Active US9993476B2 (en) 2010-05-26 2016-11-10 Substituted 5-flouro-1H-pyrazolopyridines and their use
US15/981,598 Active US10736896B2 (en) 2010-05-26 2018-05-16 Substituted 5-fluoro-1H-pyrazolopyridines and their use

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/820,667 Pending US20220409618A1 (en) 2010-05-26 2022-08-18 Substituted 5-fluoro-1h-pyrazolopyridines and their use

Country Status (45)

Country Link
US (8) US8420656B2 (en)
EP (1) EP2576547B1 (en)
JP (1) JP5896991B2 (en)
KR (1) KR101862420B1 (en)
CN (1) CN102939289B (en)
AR (1) AR081628A1 (en)
AU (1) AU2011257335B2 (en)
BR (2) BR112012030010A2 (en)
CA (1) CA2800697C (en)
CL (1) CL2012003226A1 (en)
CO (1) CO6640226A2 (en)
CR (1) CR20120596A (en)
CU (1) CU24086B1 (en)
CY (1) CY1117372T1 (en)
DE (1) DE102010021637A1 (en)
DK (1) DK2576547T3 (en)
DO (1) DOP2012000299A (en)
EA (1) EA023631B9 (en)
EC (1) ECSP12012310A (en)
ES (1) ES2567795T3 (en)
FR (1) FR21C1038I2 (en)
GT (1) GT201200313A (en)
HK (1) HK1182112A1 (en)
HR (1) HRP20160370T1 (en)
HU (2) HUE026912T2 (en)
IL (2) IL223050A (en)
JO (1) JO3199B1 (en)
LT (1) LTC2576547I2 (en)
MA (1) MA34248B1 (en)
ME (1) ME02393B (en)
MX (1) MX2012013324A (en)
MY (1) MY164356A (en)
NL (1) NL301146I2 (en)
NO (1) NO2021032I1 (en)
NZ (1) NZ603800A (en)
PE (1) PE20130402A1 (en)
PL (1) PL2576547T3 (en)
RS (1) RS54704B1 (en)
SG (1) SG185460A1 (en)
SI (1) SI2576547T1 (en)
TN (1) TN2012000549A1 (en)
TW (1) TWI516263B (en)
UA (1) UA109660C2 (en)
UY (1) UY33396A (en)
WO (1) WO2011147809A1 (en)

Families Citing this family (143)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102008063992A1 (en) * 2008-12-19 2010-09-02 Lerner, Zinoviy, Dipl.-Ing. New aliphatic substituted pyrazolopyridines and their use
DK2733142T3 (en) 2009-11-27 2018-11-19 Adverio Pharma Gmbh Process for the preparation of methyl {4,6-diamino-2- [1- (2-fluorobenzyl) -1H-pyrazolo [3,4-b] pyridin-3-yl] pyrimidin-5-yl} methylcarbamate for use as pharmaceutical active ingredient
EP2549875B1 (en) 2010-03-25 2015-05-13 Merck Sharp & Dohme Corp. Soluble guanylate cyclase activators
DE102010021637A1 (en) 2010-05-26 2011-12-01 Bayer Schering Pharma Aktiengesellschaft Substituted 5-fluoro-1H-pyrazolopyridines and their use
EA023254B1 (en) 2010-05-27 2016-05-31 Мерк Шарп Энд Домэ Корп. Soluble guanylate cyclase activators
AU2011275825A1 (en) 2010-07-09 2013-02-07 Bayer Intellectual Property Gmbh Ring-fused pyrimidines and triazines and use thereof for the treatment and/or prophylaxis of cardiovascular diseases
WO2012058132A1 (en) 2010-10-28 2012-05-03 Merck Sharp & Dohme Corp. Soluble guanylate cyclase activators
DE102010043379A1 (en) 2010-11-04 2012-05-10 Bayer Schering Pharma Aktiengesellschaft Substituted 6-fluoro-1H-pyrazolo [4,3-b] pyridines and their use
DE102010043380A1 (en) 2010-11-04 2012-05-10 Bayer Schering Pharma Aktiengesellschaft Benzyl-substituted carbamates and their use
DE102011007272A1 (en) 2011-04-13 2012-10-18 Bayer Pharma Aktiengesellschaft Branched 3-phenylpropionic acid derivatives and their use
CA2833698A1 (en) 2011-04-21 2012-10-26 Bayer Intellectual Property Gmbh Fluoroalkyl-substituted pyrazolopyridines and use thereof
EP2594270A3 (en) * 2011-11-18 2013-07-31 BIP Patents The use of sGC stimulators, sGC activators, alone and combinations with PDE5 inhibitors for the treatment of systemic sclerosis (SSc)
KR101993022B1 (en) * 2011-11-25 2019-06-25 아드베리오 파마 게엠베하 Method for producing substituted 5-fluoro-1h-pyrazolopyridines
CN102491974B (en) 2011-12-12 2013-08-07 南京药石药物研发有限公司 Method for synthesizing 1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-formamidine hydrochloride
DE102012200360A1 (en) 2012-01-11 2013-07-11 Bayer Intellectual Property Gmbh Substituted triazines and their use
DE102012200349A1 (en) 2012-01-11 2013-07-11 Bayer Intellectual Property Gmbh Substituted fused pyrimidines and triazines and their use
DE102012200352A1 (en) * 2012-01-11 2013-07-11 Bayer Intellectual Property Gmbh Substituted, fused imidazoles and pyrazoles and their use
ES2644781T3 (en) 2012-03-06 2017-11-30 Bayer Intellectual Property Gmbh Azabicycles substituted and their use
WO2013144191A1 (en) 2012-03-29 2013-10-03 Bayer Intellectual Property Gmbh Substituted 2-amino-3-cyanopyridines as inhibitors of sodium-calcium exchange and use thereof for cardiovascular diseases
US9387203B2 (en) 2012-07-20 2016-07-12 Bayer Pharma Aktiengesellschaft Substituted aminoindane- and aminotetralinecarboxylic acids and the use thereof
BR112015001211B1 (en) 2012-07-20 2020-12-15 Bayer Pharma Aktiengesellschaft 5-AMINOTETRA-HYDROQUINOLINE-2-CARBOXYLIC ACIDS, PROCESSES FOR ITS PREPARATION, ITS USES IN THE TREATMENT AND / OR PREVENTION OF DISEASES AND ITS MEDICINES
CN104822680B (en) 2012-11-30 2016-10-12 安斯泰来制药株式会社 Imidazopyridine
EP3760629A1 (en) 2013-02-21 2021-01-06 Adverio Pharma GmbH Forms of methyl {4,6-diamino-2-[1-(2-fluorobenzyl)-1h-pyrazolo[3,4-b]pyridino-3-yl]pyrimidino-5-yl}methyl carbamate
JP2016510010A (en) 2013-03-01 2016-04-04 バイエル・ファルマ・アクティエンゲゼルシャフト Trifluoromethyl-substituted ring-fused pyrimidines and uses thereof
EP3010907A1 (en) 2013-06-21 2016-04-27 Bayer Pharma Aktiengesellschaft Diaminoheteroaryl substituted pyrazoles
PE20160201A1 (en) 2013-07-10 2016-05-06 Bayer Pharma AG BENCIL-1H-PYRAZOLE [3,4-B] PYRIDINES AND THEIR USE
US10265314B2 (en) 2013-07-25 2019-04-23 Bayer Pharma Aktiengesellschaft SGC stimulators in combination with additional treatment for the therapy of cystic fibrosis
EP3046912A1 (en) 2013-09-16 2016-07-27 Bayer Pharma Aktiengesellschaft Disubstituted trifluormethyl pyrimidinones and use thereof as ccr2 antagonists
CN105793267B (en) 2013-10-07 2018-02-06 拜耳制药股份公司 Ring-type thieno uracil benzamide type and application thereof
US9783552B2 (en) 2013-12-11 2017-10-10 Merck Sharp & Dohme Corp. Soluble guanylate cyclase activators
EP3079701B1 (en) 2013-12-11 2021-08-11 Merck Sharp & Dohme Corp. Soluble guanylate cyclase activators
EP3094327A1 (en) 2014-01-13 2016-11-23 Ironwood Pharmaceuticals, Inc. USE OF sGC STIMULATORS FOR THE TREATMENT OF NEUROMUSCULAR DISORDERS
CN106458938A (en) 2014-04-03 2017-02-22 拜耳制药股份公司 Chiral 2,5-disubstituted cyclopentanecarboxylic acid derivatives and use thereof
EP3126358A1 (en) 2014-04-03 2017-02-08 Bayer Pharma Aktiengesellschaft 2,5-disubstituted cyclopentane carboxylic acids for the treatment of respiratoy tract diseases
WO2015150363A1 (en) 2014-04-03 2015-10-08 Bayer Pharma Aktiengesellschaft 2,5-disubstituted cyclopentane carboxylic acids and use thereof
WO2015187470A1 (en) 2014-06-04 2015-12-10 Merck Sharp & Dohme Corp. Imidazo-pyrazine derivatives useful as soluble guanylate cyclase activators
CN107074773A (en) 2014-09-09 2017-08-18 拜耳制药股份公司 Substituted N, the formamide of 2 biaryl quinolin 4 and application thereof
WO2016046157A1 (en) 2014-09-24 2016-03-31 Bayer Pharma Aktiengesellschaft Factor xia-inhibiting pyridobenzazepine and pyridobenzazocine derivatives
KR20170081178A (en) 2014-11-03 2017-07-11 바이엘 파마 악티엔게젤샤프트 Hydroxyalkyl-substituted phenyltriazole derivatives and uses thereof
TW201625635A (en) 2014-11-21 2016-07-16 默沙東藥廠 Triazolo-pyrazinyl derivatives useful as soluble guanylate cyclase activators
WO2016113205A1 (en) 2015-01-13 2016-07-21 Bayer Pharma Aktiengesellschaft Substituted pentafluoroethyl pyrimidinones and use thereof
UY36586A (en) 2015-03-26 2016-10-31 Bayer Pharma AG HETEROCICLILMETILTIENOURACILOS AND USE OF THE SAME
TN2017000465A1 (en) 2015-05-06 2019-04-12 Bayer Pharma AG The use of sgc stimulators, sgc activators, alone and combinations with pde5 inhibitors for the treatment of digital ulcers (du) concomitant to systemic sclerosis (ssc)
EP3303342B1 (en) 2015-05-27 2021-03-17 Merck Sharp & Dohme Corp. Imidazo-pyrazinyl derivatives useful as soluble guanylate cyclase activators
EP3310782B1 (en) 2015-05-28 2021-12-01 Merck Sharp & Dohme Corp. Imidazo-pyrazinyl derivatives useful as soluble guanylate cyclase activators
JP6849618B2 (en) 2015-07-23 2021-03-24 バイエル・ファルマ・アクティエンゲゼルシャフト Soluble guanylate cyclase (sGC) stimulants and / or activators in combination with neutral endopeptidase inhibitors (NEP inhibitors) and / or angiotensin II antagonists and their use
WO2017097671A1 (en) 2015-12-10 2017-06-15 Bayer Pharma Aktiengesellschaft Substituted perhydropyrrolo[3,4-c]pyrrole derivatives and the use of same
MX2018007040A (en) 2015-12-10 2018-08-15 Bayer Pharma AG 2-phenyl-3-(piperazinomethyl)imidazo[1,2-a]pyridine derivatives as blockers of task-1 and task-2 channels, for the treatment of sleep-related breathing disorders.
KR20180094965A (en) 2015-12-14 2018-08-24 아이언우드 파마슈티컬스, 인코포레이티드 Use of sGC Stimulants for the Treatment of Gastric Sphincter Dysfunction
CN105461715B (en) * 2015-12-15 2017-03-29 郑州大明药物科技有限公司 A kind of synthetic method of the western croak intermediate of Leo
WO2017107052A1 (en) 2015-12-22 2017-06-29 Merck Sharp & Dohme Corp. Soluble guanylate cyclase stimulators
WO2017121692A1 (en) 2016-01-15 2017-07-20 Bayer Pharma Aktiengesellschaft Substituted sulfamides and use of same
WO2017153234A1 (en) 2016-03-09 2017-09-14 Bayer Pharma Aktiengesellschaft Substituted n-cyclo-2-aryl-quinoline-4-carboxamides and use thereof
WO2017153235A1 (en) 2016-03-09 2017-09-14 Bayer Pharma Aktiengesellschaft Substituted n-cyclo-3-aryl-1-naphthamides and use thereof
WO2017153231A1 (en) 2016-03-09 2017-09-14 Bayer Pharma Aktiengesellschaft Substituted n-cyclo-2-aryl-isoquinolinone-4-carboxamides and use thereof
US10525041B2 (en) 2016-05-03 2020-01-07 Bayer Pharma Aktiengesellschaft Fluoroalkyl-substituted aryltriazole derivatives and uses thereof
US9988367B2 (en) 2016-05-03 2018-06-05 Bayer Pharma Aktiengesellschaft Amide-substituted pyridinyltriazole derivatives and uses thereof
US10800746B2 (en) 2016-05-03 2020-10-13 Bayer Pharma Aktiengesellschaft Oxoalkyl-substituted phenyltriazole derivatives and uses thereof
US20190119251A1 (en) 2016-05-03 2019-04-25 Bayer Pharma Aktiengesellschaft Amide-substituted aryltriazole derivatives and uses thereof
WO2017191114A1 (en) 2016-05-03 2017-11-09 Bayer Aktiengesellschaft Hydroxyalkyl-substituted heteroaryltriazole derivatives and uses thereof
WO2017191117A1 (en) 2016-05-03 2017-11-09 Bayer Pharma Aktiengesellschaft V1a receptor antagonists for use in the treatment of renal diseases
TW201806943A (en) 2016-05-03 2018-03-01 拜耳製藥股份有限公司 Amide-substituted phenyltriazole derivatives and uses thereof
JOP20170113B1 (en) 2016-05-09 2023-03-28 Bayer Pharma AG Substituted 5,6,7,8-tetrahydro[1,2,4]triazolo[4,3-a]pyridin-3(2H)-ones and 2,5,6,7-tetrahydro-3H-pyrrolo[2,1-c][1,2,4]triazol-3-ones and use thereof
WO2017197555A1 (en) 2016-05-16 2017-11-23 Merck Sharp & Dohme Corp. Fused pyrazine derivatives useful as soluble guanylate cyclase stimulators
US10925876B2 (en) 2016-05-18 2021-02-23 Merck Sharp & Dohme Corp. Methods for using triazolo-pyrazinyl soluble guanylate cyclase activators in fibrotic disorders
CN109689654A (en) 2016-07-11 2019-04-26 拜耳医药股份有限公司 1- pyridyl group-naphthyridines -3- benzamide type and application thereof that 7- replaces
JOP20190005A1 (en) 2016-07-20 2019-01-20 Bayer Ag Substituted diazahetero-bicyclic compounds and their use
WO2018041771A1 (en) 2016-09-02 2018-03-08 Bayer Pharma Aktiengesellschaft (1-methylcyclopropyl)methyl-substituted thienouraciles and use thereof
JOP20190045A1 (en) 2016-09-14 2019-03-14 Bayer Ag 7-substituted 1-aryl-naphthyridine-3-carboxylic acid amides and use thereof
EP3296298A1 (en) 2016-09-14 2018-03-21 Bayer Pharma Aktiengesellschaft 7-substituted 1-aryl-naphthyridin-3-carboxamides and their use
RU2019111357A (en) 2016-09-23 2020-10-23 Байер Акциенгезельшафт N3-CYCLICALLY SUBSTITUTED THYENOURACILS AND THEIR APPLICATION
CA3039734A1 (en) 2016-10-11 2018-04-19 Bayer Pharma Aktiengesellschaft Combination containing sgc stimulators and mineralocorticoid receptor antagonists
US11331308B2 (en) 2016-10-11 2022-05-17 Bayer Pharma Aktiengesellschaft Combination containing sGC activators and mineralocorticoid receptor antagonists
JOP20190080A1 (en) 2016-10-14 2019-04-11 Bayer Pharma AG Substituted 6-(1h-pyrazol-1-yl)pyrimidin-4-amine derivatives and uses thereof
US10927098B2 (en) 2016-10-20 2021-02-23 Bayer Pharma Aktiengesellschaft Hydroxyalkyl-substituted triazole derivatives and uses thereof
US20190381039A1 (en) 2016-12-13 2019-12-19 Cyclerion Therapeutics, Inc. USE OF sGC STIMULATORS FOR THE TREATMENT OF ESOPHAGEAL MOTILITY DISORDERS
JOP20190148A1 (en) 2016-12-21 2019-06-18 Bayer Pharma AG Pharmaceutical dosage forms containing task-1 and task-3 channel inhibitors, and the use of same in breathing disorder therapy
EP3338803A1 (en) 2016-12-21 2018-06-27 Bayer Pharma Aktiengesellschaft Pharmaceutical dosage forms containing inhibitors for task-1 and task-3 channels and their use in therapy of respiratory disorders
EP3338764A1 (en) 2016-12-21 2018-06-27 Bayer Pharma Aktiengesellschaft Pharmaceutical dosage forms containing inhibitors for task-1 and task-3 channels and their use in therapy of respiratory disorders
JOP20190141A1 (en) 2016-12-21 2019-06-12 Bayer Pharma AG Pharmaceutical dosage forms containing task-1 and task-3 channel inhibitors, and the use of same in breathing disorder therapy
WO2018153899A1 (en) * 2017-02-22 2018-08-30 Bayer Pharma Aktiengesellschaft Selective partial adenosine a1 receptor agonists in combination with soluble guanylyl cyclase (sgc) stimulators and/or activators
KR20190138824A (en) 2017-04-10 2019-12-16 바이엘 악티엔게젤샤프트 Substituted N-arylethyl-2-arylquinoline-4-carboxamides and uses thereof
TWI770157B (en) 2017-04-10 2022-07-11 德商拜耳廠股份有限公司 Substituted n-arylethyl-2-aminoquinoline-4-carboxamides and use thereof
CN108690016B (en) * 2017-04-11 2022-08-12 广东东阳光药业有限公司 Pyrazolopyridines and uses thereof
WO2018188590A1 (en) 2017-04-11 2018-10-18 Sunshine Lake Pharma Co., Ltd. Fluorine-substituted indazole compounds and uses thereof
JOP20190284A1 (en) 2017-06-14 2019-12-11 Bayer Pharma AG Diazabicyclic substituted imidazopyrimidines and their use for the treatment of breathing disorders
WO2018227427A1 (en) 2017-06-14 2018-12-20 Bayer Aktiengesellschaft Substituted bridged diazepane derivatives and use thereof
WO2019081291A1 (en) 2017-10-24 2019-05-02 Bayer Aktiengesellschaft Prodrugs of substituted triazole derivatives and uses thereof
US11149023B2 (en) 2017-10-24 2021-10-19 Bayer Pharma Aktiengesellschaft Substituted triazole derivatives and uses thereof
CU20200041A7 (en) * 2017-10-24 2021-03-11 Bayer Ag SUBSTITUTE IMIDAZOPYRIDINE AMIDES
US11230540B2 (en) 2017-10-24 2022-01-25 Bayer Pharma Aktiengesellschaft Substituted triazole derivatives and uses thereof
EP3700897A1 (en) 2017-10-24 2020-09-02 Bayer Pharma Aktiengesellschaft Substituted triazole derivatives and uses thereof
LT3700913T (en) 2017-10-24 2022-01-25 Bayer Aktiengesellschaft Prodrugs of substituted triazole derivatives and uses thereof
CA3079770A1 (en) 2017-10-24 2019-05-02 Bayer Pharma Aktiengesellschaft Amine substituted triazole derivatives and uses thereof
EP3700900A1 (en) 2017-10-24 2020-09-02 Bayer Aktiengesellschaft Substituted triazole derivatives and uses thereof
EP3707141B1 (en) 2017-11-07 2021-12-22 Bayer Aktiengesellschaft Substituted 2,4-dihydro-3h-1,2,4-triazol-3-ones and use of same
CA3083986A1 (en) 2017-12-01 2019-06-06 Bayer Pharma Aktiengesellschaft Method for producing (3s)-3-(4-chlor-3-{[(2s,3r)-2-(4-chlorphenyl)-4,4,4-trifluor-3-methylbutanoyl]amino}phenyl)-3-cyclo-propylpropanoic acid and the crystalline form thereof for use as a pharmaceutical ingredient
CN111712247B (en) * 2017-12-19 2024-02-09 塞科里昂医疗股份有限公司 sGC stimulators
EP3553081A1 (en) 2018-04-12 2019-10-16 Bayer Aktiengesellschaft Atrial natriuretic peptide engrafted antibodies
EP3553079A1 (en) 2018-04-12 2019-10-16 Bayer Aktiengesellschaft C-type natriuretic peptide engrafted antibodies
EP3553082A1 (en) 2018-04-12 2019-10-16 Bayer Aktiengesellschaft Brain natriuretic peptide engrafted antibodies
US20210052528A1 (en) * 2018-04-30 2021-02-25 Bayer Aktiengesellschaft The use of sgc activators and sgc stimulators for the treatment of cognitive impairment
EP3566704A1 (en) 2018-05-11 2019-11-13 Bayer Aktiengesellschaft The use of non-steroidal mineralocorticoid receptor antagonists alone or in combination for the treatment of muscular or neuromuscular diseases
AU2019269047A1 (en) 2018-05-15 2020-11-26 Bayer Aktiengesellschaft 1,3-thiazol-2-yl substituted benzamides for the treatment of diseases associated with nerve fiber sensitization
EP3793559A1 (en) 2018-05-17 2021-03-24 Bayer Aktiengesellschaft Substituted dihydropyrazolo pyrazine carboxamide derivatives
US11508483B2 (en) 2018-05-30 2022-11-22 Adverio Pharma Gmbh Method of identifying a subgroup of patients suffering from dcSSc which benefits from a treatment with sGC stimulators and sGC activators in a higher degree than a control group
WO2020014504A1 (en) 2018-07-11 2020-01-16 Cyclerion Therapeutics, Inc. USE OF sGC STIMULATORS FOR THE TREATMENT OF MITOCHONRIAL DISORDERS
EP3826619A1 (en) 2018-07-24 2021-06-02 Bayer Aktiengesellschaft Pharmaceutical dosage form which can be administered orally and has modified release
SG11202100369WA (en) 2018-07-24 2021-02-25 Bayer Ag Pharmaceutical dosage form which can be administered orally and has modified release
US10905667B2 (en) 2018-07-24 2021-02-02 Bayer Pharma Aktiengesellschaft Orally administrable modified-release pharmaceutical dosage form
EP3886806A1 (en) 2018-11-27 2021-10-06 Bayer Aktiengesellschaft Process for producing pharmaceutical dosage forms containing task-1 and task-3 channel inhibitors, and the use of same in breathing disorder therapy
CA3123360A1 (en) 2018-12-17 2020-06-25 Adverio Pharma Gmbh Methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1h-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate active compound product having improved properties, production and formulation thereof
WO2020164008A1 (en) 2019-02-13 2020-08-20 Bayer Aktiengesellschaft Process for the preparation of porous microparticles
WO2020216669A1 (en) 2019-04-23 2020-10-29 Bayer Aktiengesellschaft Phenyl-substituted imidazopyridine amides and use thereof
AU2020268578A1 (en) 2019-05-07 2022-02-03 Bayer Aktiengesellschaft MASP inhibitory compounds and uses thereof
WO2020245342A1 (en) 2019-06-07 2020-12-10 Bayer Aktiengesellschaft The use of sgc activators for the treatment of ophthalmologic diseases
CA3160035A1 (en) 2019-11-06 2021-05-14 Bayer Aktiengesellschaft Inhibitors of adrenoreceptor adrac2
WO2021094209A1 (en) 2019-11-12 2021-05-20 Bayer Aktiengesellschaft Substituted pyrrolo triazine carboxamide derivatives as prostaglandin ep3 receptor antagonists
WO2021094210A1 (en) 2019-11-12 2021-05-20 Bayer Aktiengesellschaft Substituted pyrazine carboxamide derivatives as prostaglandin ep3 receptor antagonists
WO2021094208A1 (en) 2019-11-12 2021-05-20 Bayer Aktiengesellschaft Substituted imidazo pyrimidine ep3 antagonists
EP3822265A1 (en) 2019-11-15 2021-05-19 Bayer AG Substituted hydantoinamides as adamts7 antagonists
EP3822268A1 (en) 2019-11-15 2021-05-19 Bayer Aktiengesellschaft Substituted hydantoinamides as adamts7 antagonists
WO2021156223A1 (en) 2020-02-03 2021-08-12 Adverio Pharma Gmbh Nanoformulations of methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1h-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate
EP4106741A1 (en) 2020-02-21 2022-12-28 Universiteit Maastricht Use of a soluble guanylate cyclase (sgc) stimulator or of a combination of a sgc stimulator and an sgc activator for conditions wherein the heme group of sgc is oxidized or wherein sgc is deficient in heme
EP4110396A1 (en) 2020-02-26 2023-01-04 Universiteit Maastricht Therapeutic combination for the treatment of brain ischemia and said therapeutic combination for use in the treatment of brain ischemia
WO2021195403A1 (en) 2020-03-26 2021-09-30 Cyclerion Therapeutics, Inc. Deuterated sgc stimulators
EP3925953A1 (en) 2020-06-16 2021-12-22 Adverio Pharma GmbH Process for preparing methyl {4,6-diamino-2-[5-fluoro-1-(2-fluorobenzyl)-1h-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate
EP4011904A1 (en) 2020-12-14 2022-06-15 Bayer Aktiengesellschaft Masp inhibitory compounds and uses thereof
US20240010684A1 (en) 2020-11-04 2024-01-11 Bayer Aktiengesellschaft Masp inhibitory compounds and uses thereof
WO2022112213A1 (en) 2020-11-30 2022-06-02 Bayer Aktiengesellschaft Crystalline forms of 3-[[3-(4-chlorophenyl)-5-oxo-4-((2s)-3,3,3-trifluoro- 2-hydroxypropyl)-4,5-dihydro-1h-1,2,4-triazol-1-yl]methyl]-1-[3- (trifluoromethyl)pyridin-2-yl]-1h-1,2,4-triazole-5-carboxamide
WO2022122914A1 (en) 2020-12-10 2022-06-16 Bayer Aktiengesellschaft Substituted pyrazolo piperidine carboxylic acids
EP4011873A1 (en) 2020-12-10 2022-06-15 Bayer Aktiengesellschaft Substituted pyrazolo piperidine carboxylic acids
EP4011874A1 (en) 2020-12-10 2022-06-15 Bayer Aktiengesellschaft Substituted pyrazolo piperidine carboxylic acids
CA3204595A1 (en) 2020-12-10 2022-06-16 Bayer Aktiengesellschaft Substituted pyrazolyl piperidine carboxylic acids
KR20230118143A (en) 2020-12-10 2023-08-10 바이엘 악티엔게젤샤프트 Use of sGC activators for the treatment of ophthalmic diseases
TW202309038A (en) 2021-04-20 2023-03-01 美商賽克瑞恩醫療公司 Sgc stimulators
TW202308634A (en) 2021-04-20 2023-03-01 美商賽克瑞恩醫療公司 Treatment of cns diseases with sgc stimulators
WO2023034364A1 (en) 2021-08-31 2023-03-09 Teva Pharmaceuticals International Gmbh Solid state forms of vericiguat and process for preparation thereof
WO2023126437A1 (en) 2021-12-29 2023-07-06 Bayer Aktiengesellschaft Treatment of cardiopulmonary disorders
WO2023126436A1 (en) 2021-12-29 2023-07-06 Bayer Aktiengesellschaft Process for preparing (5s)-{[2-(4-carboxyphenyl)ethyl] |2-(2-{|3-chloro-4'-(trifluoromethyl)biphenyl-4- yl]methoxy}phenyl)ethyl]aminol-5,6,7,8-tetrahydroquinoline-2-carboxylic acid and its crystalline forms for use as pharmaceutically active compound
WO2023126438A1 (en) 2021-12-29 2023-07-06 Bayer Aktiengesellschaft Pharmaceutical dry powder inhalation formulation
ES2956054A1 (en) * 2022-05-03 2023-12-12 Moehs Iberica Sl 2-(5-FLUORO-1-(2-FLUOROBENZYL)-1H-PYRAZOLO[3,4-b]PYRIDIN-3-IL)-5-NITROSOPYRIMIDIN-4,6-DIAMINE OR A SALT THEREOF, PROCEDURE FOR ITS PREPARATION AND ITS USE IN THE SYNTHESIS OF VERICIGUAT (Machine-translation by Google Translate, not legally binding)
WO2023237577A1 (en) 2022-06-09 2023-12-14 Bayer Aktiengesellschaft Soluble guanylate cyclase activators for use in the treatment of heart failure with preserved ejection fraction in women

Citations (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0463756A1 (en) 1990-06-20 1992-01-02 Pfizer Limited Pyrazolopyrimidinone antianginal agents
EP0463754A1 (en) 1990-06-25 1992-01-02 Rohm And Haas Company Stabilization of glutarimide polymers
CA2127624A1 (en) 1993-07-13 1995-01-14 Barry D. G. Haylor Herbicides
US5393761A (en) 1990-07-30 1995-02-28 H. Lundbeck A/S 3-arylindole compounds
US6008243A (en) 1996-10-24 1999-12-28 Agouron Pharmaceuticals, Inc. Metalloproteinase inhibitors, pharmaceutical compositions containing them, and their use
WO2000006567A1 (en) 1998-07-29 2000-02-10 Bayer Aktiengesellschaft 3-(4-amino-5-ethylpyrimidine-2-yl)-1-(2-fluorobenzyl)-1h-pyrazolo[3,4-b]pyridine
WO2000021954A1 (en) 1998-10-09 2000-04-20 Bayer Aktiengesellschaft Novel heterocyclyl-methyl-substituted pyrazoles
US6090818A (en) 1996-12-13 2000-07-18 Sanofi-Synthelabo Indolin-2-one derivatives, method for preparing them and pharmaceutical compositions containing them
US6166027A (en) 1996-10-14 2000-12-26 Bayer Aktiengesellschaft Heterocyclylmethyl-substituted pyrazole derivatives and their use for treating cardiovascular diseases
US6180656B1 (en) 1996-10-14 2001-01-30 Bayer Aktiengesellschaft Use of condensated (hetaryl-substituted) 1-benzal-3-pyrazol derivates for treating special diseases of the cardiovascular and the central nervous systems
US6362178B1 (en) 1997-11-12 2002-03-26 Bayer Aktiengesellschaft 2-phenyl substituted imidazotriazinones as phosphodiesterase inhibitors
US6451805B1 (en) 1997-11-14 2002-09-17 Bayer Aktiengesellschaft Substituted pyrazole derivatives for the treatment of cardiocirculatory diseases
US6469012B1 (en) 1993-06-09 2002-10-22 Pfizer Inc Pyrazolopyrimidinones for the treatment of impotence
US6576651B2 (en) 2001-01-25 2003-06-10 Bristol-Myers Squibb Company Pharmaceutical compositions, dosage forms and methods for oral administration of epothilones
WO2003076408A2 (en) 2002-03-08 2003-09-18 Abbott Laboratories Indazole derivatives that are activators of soluble guanylate cyclase
US6693102B2 (en) 2000-11-22 2004-02-17 Bayer Aktiengesellschaft Pyridine-substituted pyrazolopyridine derivatives
WO2004031187A1 (en) 2002-09-16 2004-04-15 Bayer Healthcare Ag Pyridine-substituted pyrazolopyridine derivative
US6743798B1 (en) 1998-07-29 2004-06-01 Bayer Aktiengesellschaft Substituted pyrazole derivatives condensed with six-membered heterocyclic rings
US6784179B2 (en) 1994-01-21 2004-08-31 Icos Corporation Tetracyclic derivatives, process of preparation and use
US20040235863A1 (en) 2001-07-04 2004-11-25 Achim Feurer Morpholine-bridged pyrazolopyridine derivatives
US6833364B1 (en) 1998-07-29 2004-12-21 Bayer Healthcare Ag Substituted pyrazole derivatives
CN1613849A (en) 2003-11-03 2005-05-11 上海药明康德新药开发有限公司 Preparation of 2-Cl-5-F-nicotinate and nicotonic acid
US6903089B1 (en) 2000-11-22 2005-06-07 Bayer Aktiengesellschaft Lactam-substituted pyrazolopyridine derivatives
US6919345B2 (en) 2001-05-11 2005-07-19 Bayer Aktiengesellschaft Sulfonate substituted pyrazol pyridine derivatives
CA2577420A1 (en) 2004-08-20 2006-03-02 University Of Kansas Lonidamine analogues and their use in male contraception and cancer treatment
US20060167016A1 (en) 2002-07-18 2006-07-27 Achim Feurer 2,5-disubstituted pyrimidine derivatives
US7105523B2 (en) 2000-11-22 2006-09-12 Bayer Aktiengeselischaft Carbamate-substituted pyrazolopyridine-derivatives
US7115599B2 (en) 2000-11-22 2006-10-03 Bayer Aktiengesellschaft Sulfonamide-substituted pyrazolopyridine compounds
US7135474B2 (en) 2002-05-17 2006-11-14 Bayer Healthcare Ag Derivatives of 2-(1-benzyl-1H-pyrazolo(3,4-B)pyridine-3-yl)-5(-4-pyridinyl)l-4-pyrimidinamines and the use thereof as quanylate cyclase stimulators
US20070015841A1 (en) 2002-02-15 2007-01-18 Transform Pharmaceuticals, Inc. Pharmaceutical propylene glycol solvate compositions
WO2007009607A1 (en) 2005-07-18 2007-01-25 Bayer Healthcare Ag Novel use of activators and stimulators of soluble guanylate cyclase for the prevention or treatment of renal disorders
US7173037B2 (en) 2002-05-08 2007-02-06 Bayer Healthcare Ag Carbamate-substituted pyrazolopyridines
US7176220B2 (en) 2002-11-20 2007-02-13 Japan Tobacco Inc. 4-oxoquinoline compound and use thereof as pharmaceutical agent
US20070072924A1 (en) 2003-10-31 2007-03-29 Graeme Semple Tetrazole derivatives and methods of treatment of metabolic-related disorders thereof
US7226941B2 (en) 2003-06-30 2007-06-05 Hif Bio, Inc. Compound for treating angiogenesis
US20070213332A1 (en) 2004-02-20 2007-09-13 Frank Burkamp Prodrugs Of Substituted Amino Heterobicycles Which Modulate The Function Of The Vanilloid-1 Receptor (Vr1)
US20070225299A1 (en) 2003-11-06 2007-09-27 Bayer Healthcare Ag Novel Combination Containing a Stimulator of Soluble Guanylate Cyclase and a Lipid-Lowering Substance
US20080138444A1 (en) 2004-10-05 2008-06-12 Bayer Healthcare Ag Method For Treating Bronchoconstriction and Pulmonary Vaso-Constriction
US7410973B2 (en) 2002-07-18 2008-08-12 Bayer Healthcare Ag 4-amino-substituted pyrimidine derivatives
US7427617B2 (en) 2002-09-26 2008-09-23 Bayer Healthcare Ag Morpoline-bridged indazole derivatives
WO2009000832A2 (en) 2007-06-25 2008-12-31 Boehringer Ingelheim International Gmbh Chemical compounds
US20090215769A1 (en) 2005-07-16 2009-08-27 Bayer Healthcare Ag Use of soluble guanylate cyclase activators for the treatment of Raynaud's Phenomenon
US7666867B2 (en) 2001-10-26 2010-02-23 University Of Connecticut Heteroindanes: a new class of potent cannabimimetic ligands
US20100113507A1 (en) 2006-05-09 2010-05-06 Bayer Healthcare Ag 3-Tetrazolyl Indazoles, 3-Tetrazolyl Pyrazolopyridines, and use Thereof
US7790761B2 (en) 2000-02-04 2010-09-07 University College London Blockade of voltage dependent sodium channels
US20110183999A1 (en) 2009-11-27 2011-07-28 Bayer Schering Pharma Aktiengesellschaft Novel polymorphic forms of methyl carbamate
US20110224197A1 (en) 2008-03-10 2011-09-15 Vertex Pharmaceuticals Incorporated Pyrimidines and pyridines useful as inhibitors of protein kinases
US20120029002A1 (en) 2009-01-09 2012-02-02 Bayer Schering Pharma Aktiengesellschaft Benzimidazole and pyrazolopyridine derivatives for treating and/or preventing cardiovascular diseases
CA2809911A1 (en) 2010-09-03 2012-03-08 Markus Follmann Bicyclic aza heterocycles, and use thereof
US8242272B2 (en) 2007-07-31 2012-08-14 Vertex Pharmaceuticals Inc. Process for preparing 5-fluoro-1H-pyrazolo [3,4-B] pyridin-3-amine and derivatives thereof
US8309551B2 (en) 2006-09-15 2012-11-13 Bayer Intellectual Property Gmbh Pyrazolopyridine, indazole, imidazopyridine, imidazopyrimidine, pyrazolopyrazine and pyrazolopyridine derivatives as stimulators of guanylate cyclase for cardiovascular disorders
US20120309724A1 (en) 2011-05-31 2012-12-06 Theravance, Inc. Neprilysin inhibitors
US8334291B2 (en) 2008-12-19 2012-12-18 Bayer Intellectual Property Gmbh Aliphatically substituted pyrazolopyridines, and the use thereof
US20130053393A1 (en) 2009-12-18 2013-02-28 Exodos Life Sciences Limited Partnership Methods and compositions for treating peripheral vascular disease
US8420656B2 (en) 2010-05-26 2013-04-16 Bayer Intellectual Property Gmbh Substituted 5-fluoro-1H-pyrazolopyridines and their use
US20130178475A1 (en) 2010-03-17 2013-07-11 Ironwood Pharmaceuticals, Inc. sGC STIMULATORS
US8492544B2 (en) 2009-11-27 2013-07-23 Bayer Intellectual Property Gmbh Process for preparing methyl {4,6-diamino-2-[1-(2-fluorobenzyl)-1H-pyrazolo[3,4-B]pyridin-3-yl]pyrimidin-5-yl}Methylcarbamate and its purification for use as pharmaceutically active compound
US8501945B2 (en) 2009-11-27 2013-08-06 Bayer Intellectual Property Gmbh Process for preparing methyl {4,6-diamino-2-[1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate and its purification for use as pharmaceutically active compound
US20130211090A1 (en) 2010-09-03 2013-08-15 Bayer Intellectual Property Gmbh Method for the production of 5-fluoro-1h-pyrazolo[3,4-b]pyridine-3-carbonitrile
US20130267548A1 (en) 2010-11-04 2013-10-10 Bayer Intellectual Property Gmbh Substituted 6-fluoro-1h-pyrazolo[4,3-b]pyridines and use thereof
US8802847B2 (en) 2011-11-25 2014-08-12 Bayer Intellectual Property Gmbh Process for preparing substituted 5-fluoro-1H-pyrazolopyridines
US8859569B2 (en) 2011-09-02 2014-10-14 Bayer Pharma Aktiengesellschaft Substituted annellated pyrimidines and use thereof
US8927557B2 (en) 2009-08-26 2015-01-06 Takeda Gmbh Methylpyrrolopyrimidinecarboxamides
US20150065533A1 (en) 2010-11-04 2015-03-05 Bayer Intellectual Property Gmbh Benzyl-substituted carbamates and use thereof
US9023849B2 (en) 2012-01-11 2015-05-05 Bayer Intellectual Property Gmbh Substituted fused imidazoles and pyrazoles and use thereof
US9216978B2 (en) 2010-07-09 2015-12-22 Bayer Intellectual Property Gmbh Ring-fused pyrimidines and triazines and use thereof for the treatment and/or prophylaxis of cardiovascular diseases
US9365574B2 (en) 2010-05-27 2016-06-14 Merck Sharp & Dohme Corp. Soluble guanylate cyclase activators

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH01198481A (en) * 1988-02-01 1989-08-10 Canon Inc Formation of deposited film by microwave plasma cvd
GR1004350B (en) * 2002-03-29 2003-09-26 Inhaler for dry powder
EP1626045B1 (en) 2003-05-09 2010-09-08 Asahi Glass Company, Limited Processes for producing 3-substituted 2-chloro-5-fluoropyridine or salt thereof
RS54261B1 (en) * 2010-05-26 2016-02-29 Adverio Pharma Gmbh The use of sgc stimulators, sgc activators, alone and combinations with pde5 inhibitors for the treatment of systemic sclerosis (ssc)

Patent Citations (80)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0463756A1 (en) 1990-06-20 1992-01-02 Pfizer Limited Pyrazolopyrimidinone antianginal agents
EP0463754A1 (en) 1990-06-25 1992-01-02 Rohm And Haas Company Stabilization of glutarimide polymers
US5393761A (en) 1990-07-30 1995-02-28 H. Lundbeck A/S 3-arylindole compounds
US6469012B1 (en) 1993-06-09 2002-10-22 Pfizer Inc Pyrazolopyrimidinones for the treatment of impotence
CA2127624A1 (en) 1993-07-13 1995-01-14 Barry D. G. Haylor Herbicides
EP0634413A1 (en) 1993-07-13 1995-01-18 Rhone Poulenc Agriculture Ltd. Herbicides
US6784179B2 (en) 1994-01-21 2004-08-31 Icos Corporation Tetracyclic derivatives, process of preparation and use
US6166027A (en) 1996-10-14 2000-12-26 Bayer Aktiengesellschaft Heterocyclylmethyl-substituted pyrazole derivatives and their use for treating cardiovascular diseases
US6180656B1 (en) 1996-10-14 2001-01-30 Bayer Aktiengesellschaft Use of condensated (hetaryl-substituted) 1-benzal-3-pyrazol derivates for treating special diseases of the cardiovascular and the central nervous systems
US6008243A (en) 1996-10-24 1999-12-28 Agouron Pharmaceuticals, Inc. Metalloproteinase inhibitors, pharmaceutical compositions containing them, and their use
US6090818A (en) 1996-12-13 2000-07-18 Sanofi-Synthelabo Indolin-2-one derivatives, method for preparing them and pharmaceutical compositions containing them
US6362178B1 (en) 1997-11-12 2002-03-26 Bayer Aktiengesellschaft 2-phenyl substituted imidazotriazinones as phosphodiesterase inhibitors
US6451805B1 (en) 1997-11-14 2002-09-17 Bayer Aktiengesellschaft Substituted pyrazole derivatives for the treatment of cardiocirculatory diseases
WO2000006567A1 (en) 1998-07-29 2000-02-10 Bayer Aktiengesellschaft 3-(4-amino-5-ethylpyrimidine-2-yl)-1-(2-fluorobenzyl)-1h-pyrazolo[3,4-b]pyridine
US6743798B1 (en) 1998-07-29 2004-06-01 Bayer Aktiengesellschaft Substituted pyrazole derivatives condensed with six-membered heterocyclic rings
US6833364B1 (en) 1998-07-29 2004-12-21 Bayer Healthcare Ag Substituted pyrazole derivatives
CA2346698A1 (en) 1998-10-09 2000-04-20 Bayer Aktiengesellschaft Novel heterocyclyl-methyl-substituted pyrazoles
WO2000021954A1 (en) 1998-10-09 2000-04-20 Bayer Aktiengesellschaft Novel heterocyclyl-methyl-substituted pyrazoles
US7790761B2 (en) 2000-02-04 2010-09-07 University College London Blockade of voltage dependent sodium channels
US6693102B2 (en) 2000-11-22 2004-02-17 Bayer Aktiengesellschaft Pyridine-substituted pyrazolopyridine derivatives
US7105523B2 (en) 2000-11-22 2006-09-12 Bayer Aktiengeselischaft Carbamate-substituted pyrazolopyridine-derivatives
US6903089B1 (en) 2000-11-22 2005-06-07 Bayer Aktiengesellschaft Lactam-substituted pyrazolopyridine derivatives
US7115599B2 (en) 2000-11-22 2006-10-03 Bayer Aktiengesellschaft Sulfonamide-substituted pyrazolopyridine compounds
US6576651B2 (en) 2001-01-25 2003-06-10 Bristol-Myers Squibb Company Pharmaceutical compositions, dosage forms and methods for oral administration of epothilones
US6919345B2 (en) 2001-05-11 2005-07-19 Bayer Aktiengesellschaft Sulfonate substituted pyrazol pyridine derivatives
US20040235863A1 (en) 2001-07-04 2004-11-25 Achim Feurer Morpholine-bridged pyrazolopyridine derivatives
US7666867B2 (en) 2001-10-26 2010-02-23 University Of Connecticut Heteroindanes: a new class of potent cannabimimetic ligands
US20070015841A1 (en) 2002-02-15 2007-01-18 Transform Pharmaceuticals, Inc. Pharmaceutical propylene glycol solvate compositions
US7790905B2 (en) 2002-02-15 2010-09-07 Mcneil-Ppc, Inc. Pharmaceutical propylene glycol solvate compositions
WO2003076408A2 (en) 2002-03-08 2003-09-18 Abbott Laboratories Indazole derivatives that are activators of soluble guanylate cyclase
US7173037B2 (en) 2002-05-08 2007-02-06 Bayer Healthcare Ag Carbamate-substituted pyrazolopyridines
US7135474B2 (en) 2002-05-17 2006-11-14 Bayer Healthcare Ag Derivatives of 2-(1-benzyl-1H-pyrazolo(3,4-B)pyridine-3-yl)-5(-4-pyridinyl)l-4-pyrimidinamines and the use thereof as quanylate cyclase stimulators
US7091198B1 (en) 2002-07-18 2006-08-15 Bayer Healthcare Ag 2,5-disubstituted pyrimidine derivatives
US20060167016A1 (en) 2002-07-18 2006-07-27 Achim Feurer 2,5-disubstituted pyrimidine derivatives
US7410973B2 (en) 2002-07-18 2008-08-12 Bayer Healthcare Ag 4-amino-substituted pyrimidine derivatives
WO2004031187A1 (en) 2002-09-16 2004-04-15 Bayer Healthcare Ag Pyridine-substituted pyrazolopyridine derivative
US7427617B2 (en) 2002-09-26 2008-09-23 Bayer Healthcare Ag Morpoline-bridged indazole derivatives
US7176220B2 (en) 2002-11-20 2007-02-13 Japan Tobacco Inc. 4-oxoquinoline compound and use thereof as pharmaceutical agent
US7226941B2 (en) 2003-06-30 2007-06-05 Hif Bio, Inc. Compound for treating angiogenesis
US20070072924A1 (en) 2003-10-31 2007-03-29 Graeme Semple Tetrazole derivatives and methods of treatment of metabolic-related disorders thereof
CN1613849A (en) 2003-11-03 2005-05-11 上海药明康德新药开发有限公司 Preparation of 2-Cl-5-F-nicotinate and nicotonic acid
US20070225299A1 (en) 2003-11-06 2007-09-27 Bayer Healthcare Ag Novel Combination Containing a Stimulator of Soluble Guanylate Cyclase and a Lipid-Lowering Substance
US20070213332A1 (en) 2004-02-20 2007-09-13 Frank Burkamp Prodrugs Of Substituted Amino Heterobicycles Which Modulate The Function Of The Vanilloid-1 Receptor (Vr1)
US7514463B2 (en) 2004-08-20 2009-04-07 University Of Kansas Lonidamine analogues and their use in male contraception and cancer treatment
CA2577420A1 (en) 2004-08-20 2006-03-02 University Of Kansas Lonidamine analogues and their use in male contraception and cancer treatment
US20080138444A1 (en) 2004-10-05 2008-06-12 Bayer Healthcare Ag Method For Treating Bronchoconstriction and Pulmonary Vaso-Constriction
US20090215769A1 (en) 2005-07-16 2009-08-27 Bayer Healthcare Ag Use of soluble guanylate cyclase activators for the treatment of Raynaud's Phenomenon
WO2007009607A1 (en) 2005-07-18 2007-01-25 Bayer Healthcare Ag Novel use of activators and stimulators of soluble guanylate cyclase for the prevention or treatment of renal disorders
US20100113507A1 (en) 2006-05-09 2010-05-06 Bayer Healthcare Ag 3-Tetrazolyl Indazoles, 3-Tetrazolyl Pyrazolopyridines, and use Thereof
US8309551B2 (en) 2006-09-15 2012-11-13 Bayer Intellectual Property Gmbh Pyrazolopyridine, indazole, imidazopyridine, imidazopyrimidine, pyrazolopyrazine and pyrazolopyridine derivatives as stimulators of guanylate cyclase for cardiovascular disorders
WO2009000832A2 (en) 2007-06-25 2008-12-31 Boehringer Ingelheim International Gmbh Chemical compounds
US8242272B2 (en) 2007-07-31 2012-08-14 Vertex Pharmaceuticals Inc. Process for preparing 5-fluoro-1H-pyrazolo [3,4-B] pyridin-3-amine and derivatives thereof
US20110224197A1 (en) 2008-03-10 2011-09-15 Vertex Pharmaceuticals Incorporated Pyrimidines and pyridines useful as inhibitors of protein kinases
US8334291B2 (en) 2008-12-19 2012-12-18 Bayer Intellectual Property Gmbh Aliphatically substituted pyrazolopyridines, and the use thereof
US20120029002A1 (en) 2009-01-09 2012-02-02 Bayer Schering Pharma Aktiengesellschaft Benzimidazole and pyrazolopyridine derivatives for treating and/or preventing cardiovascular diseases
US8927557B2 (en) 2009-08-26 2015-01-06 Takeda Gmbh Methylpyrrolopyrimidinecarboxamides
US8492544B2 (en) 2009-11-27 2013-07-23 Bayer Intellectual Property Gmbh Process for preparing methyl {4,6-diamino-2-[1-(2-fluorobenzyl)-1H-pyrazolo[3,4-B]pyridin-3-yl]pyrimidin-5-yl}Methylcarbamate and its purification for use as pharmaceutically active compound
US8853398B2 (en) 2009-11-27 2014-10-07 Bayer Intellectual Property Gmbh Process for preparing methyl {4,6-diamino-2-[1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b] pyridin-3-yl]pyrimidin-5-yl}methylcarbamate and its purification for use as pharmaceutically active compound
US8501945B2 (en) 2009-11-27 2013-08-06 Bayer Intellectual Property Gmbh Process for preparing methyl {4,6-diamino-2-[1-(2-fluorobenzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]pyrimidin-5-yl}carbamate and its purification for use as pharmaceutically active compound
US20110183999A1 (en) 2009-11-27 2011-07-28 Bayer Schering Pharma Aktiengesellschaft Novel polymorphic forms of methyl carbamate
US20130053393A1 (en) 2009-12-18 2013-02-28 Exodos Life Sciences Limited Partnership Methods and compositions for treating peripheral vascular disease
US20130178475A1 (en) 2010-03-17 2013-07-11 Ironwood Pharmaceuticals, Inc. sGC STIMULATORS
US8921377B2 (en) 2010-05-26 2014-12-30 Bayer Intellectual Property Gmbh Substituted 5-fluoro-1H-pyrazolopyridines and their use
US9993476B2 (en) * 2010-05-26 2018-06-12 Adverio Pharma Gmbh Substituted 5-flouro-1H-pyrazolopyridines and their use
US9266885B2 (en) 2010-05-26 2016-02-23 Adverio Pharma Gmbh Substituted 5-fluoro-1H-pyrazolopyridines and their use
US8420656B2 (en) 2010-05-26 2013-04-16 Bayer Intellectual Property Gmbh Substituted 5-fluoro-1H-pyrazolopyridines and their use
US9365574B2 (en) 2010-05-27 2016-06-14 Merck Sharp & Dohme Corp. Soluble guanylate cyclase activators
US9216978B2 (en) 2010-07-09 2015-12-22 Bayer Intellectual Property Gmbh Ring-fused pyrimidines and triazines and use thereof for the treatment and/or prophylaxis of cardiovascular diseases
US9096592B2 (en) 2010-09-03 2015-08-04 Bayer Intellectual Property Gmbh Bicyclic aza heterocycles, and use thereof
CA2809911A1 (en) 2010-09-03 2012-03-08 Markus Follmann Bicyclic aza heterocycles, and use thereof
US20130211090A1 (en) 2010-09-03 2013-08-15 Bayer Intellectual Property Gmbh Method for the production of 5-fluoro-1h-pyrazolo[3,4-b]pyridine-3-carbonitrile
US20150065533A1 (en) 2010-11-04 2015-03-05 Bayer Intellectual Property Gmbh Benzyl-substituted carbamates and use thereof
US9090609B2 (en) 2010-11-04 2015-07-28 Bayer Intellectual Property Gmbh Benzyl-substituted carbamates and use thereof
US20130267548A1 (en) 2010-11-04 2013-10-10 Bayer Intellectual Property Gmbh Substituted 6-fluoro-1h-pyrazolo[4,3-b]pyridines and use thereof
US20120309724A1 (en) 2011-05-31 2012-12-06 Theravance, Inc. Neprilysin inhibitors
US8859569B2 (en) 2011-09-02 2014-10-14 Bayer Pharma Aktiengesellschaft Substituted annellated pyrimidines and use thereof
US20140315926A1 (en) 2011-11-25 2014-10-23 Bayer Intellectual Property Gmbh Process for preparing substituted 5-fluoro-1H-pyrazolopyridines
US9150573B2 (en) 2011-11-25 2015-10-06 Adverio Pharma Gmbh Process for preparing substituted 5-fluoro-1H-pyrazolopyridines
US8802847B2 (en) 2011-11-25 2014-08-12 Bayer Intellectual Property Gmbh Process for preparing substituted 5-fluoro-1H-pyrazolopyridines
US9023849B2 (en) 2012-01-11 2015-05-05 Bayer Intellectual Property Gmbh Substituted fused imidazoles and pyrazoles and use thereof

Non-Patent Citations (66)

* Cited by examiner, † Cited by third party
Title
Armstrong, P.W. et al., "Vericiguat in Patients with Heart Failure and Reduced Ejection Fraction," N Engl J Med 2020, 382(20), 1883-1893.
Barraclough et al.,"Mono-aroylation of 2,3- and 3,4-Diaminopyridine and 4,5-Diaminopyrimidine, and Syntheses of Putative Inotrope/β-Adrenoceptor Antagonists," J. Chem. Res., 1996, 9: 2316-2335.
Becker et al.,"NO-Independent Regulatory Site of Direct sGC Stimulators like YC-1 and BAY 41-2272," BMC Pharmacology, 2001, 1: 13.
Boerrigter, G, et al. "Cardiorenal and humoral properties of a novel direct soluble guanylate cyclase stimulator BAY 41-2272 in experimental congestive heart failure," Circulation, 2003, 686-689, 107.
Breitenstein, S. et al., "Novel sGC Stimulators and sGC Activators for the Treatment of Heart Failure" in J. Bauersachs et al. (eds.), Heart Failure, Handbook of Experimental Pharmacology, 2016, 225-247, 243.
Britain's publication, crystalline and pharmaceutical composition, 1999, pp. 348-361.
Cavalieri et al.,"A Synthesis of Adenine: The Incorporation of Isotopes of Nitrogen and Carbon," J. Am. Chem. Soc., Feb. 1949, 71:533-536.
Cheng et al., "Potential Purine Antagonists VII. Synthesis of 6-Alkylpyrazolo-[3,4-d]pyrimidines," J. Org. Chem., 1958, 23:191-200.
Corsi et al., "1-Halobanzyl-1H-indazole-3-carboxylic acids. A New Class of Antispermatogenic Agents," Journal of Medicinal Chemistry, 1976, 19(6):778-83.
Costell, M. et al. "Comparison of soluble guanylate cyclase stimulators and activators in models of cardiovascular disease associated with oxidative stress," Frontiers in Pharmacology, Jul. 2012, 1-14, vol. 3, Article 18.
Evans et al., "The Preparation of 4-Amino- and Other Pteridines," J. of Chem. Soc., 1956, pp. 4106-4113.
Evgenov et al., "Inhaled Agonists of Soluble Guanylate Cyclase Induce Selective Pulmonary Vasodilation," Am J respir Crit Care Med, vol. 176, pp. 1138-1145, 2007.
Evgenov et al., "NO-independent stimulators and activators of soluble guanylate cyclase: discovery and therapeutic potential," Nat. Rev. Drug. Disc, Sep. 2006, 5:755-768.
Follmann, M. et al. "Discovery of the Soluble Guanylate Cyclase Stimulator Vericiguat (BAY 1021189) for the Treatment of Chronic Heart Failure," J. Med. Chem., 2017, 5146-5161, 60.
Funabiki et al., "Fluoride Ion-Promoted Reaction of Polyfluoro-1-propenyl p-Toluenesulfonate with Amines. Highly Efficient and General Access to (Z)-α-Fluoro-β-amino Acrylaldehydes," Chemistry Letters 1994, vol. 6, 1075-1078.
Geschka, S. et al. "Soluble Guanylate Cyclase Stimulation Prevents Fibrotic Tissue Remodeling and Improves Survival in Salt-Sensitive Dahl Rats," PLoS ONE Jul. 2011, 1-10, vol. 6, Issue 7, e21853.
Gheorghiade, M., et al. "Effect of Vericiguat, a Soluble Guanylate Cyclase Stimulator, on Natriuretic Peptide Levels in Patients With Worsening Chronic Heart Failure and Reduced Ejection Fraction," JAMA 2015, 2251-2262, 314(21).
Ghofrani et al., "Soluble Guanylate Cyclase Stimulation: An emerging option in Pulmonary Hypertension Therapy" European Respiratory Review, vol. 18, No. 111, pp. 35-41, 2009.
Glass et al., "Stimulation of Human Platelet Guanylate Cyclase by Fatty Acids," J. Biol. Chem., Feb. 25, 1977, 252 (4): 1279-1285.
Gore, J. et al., "Oral sildenafil for the treatment of Raynaud's phenomenon and digital ulcers secondary to systemic sclerosis," Ann Rheum Dis., 2005, 64, p. 1387.
Greene et al., "The cGMP Signaling Pathway as a Therapeutic Target in Heart Failure With Preserved Ejection Fraction," Journal of the American Heart Association, Dec. 11, 2013.
Guillory, "Generation of Polymorphs, Hydrates, Solvates, and Amorphous Solids," Polymorphism in Pharmaceutical solids p. 183-220, 188 (H.G. Brittain ed., 1999).
Hajos et al., "Product Class 5: Azaindolizines with Two Nitrogen Atoms in the Five-Membered Ring," Science of Synthesis, 2002, 12:613-678.
Hassan et al., "Aryl-Aryl Bond Formation One Century after the Discovery of the Ullmann Reaction," Chem. Rev. 2002, 102: 1359-1469.
Hughes,"Progress in the Mitsunobu Reaction. A Review," Org. Prep. Procedures Int., 1996, 28(2):127-164.
Hughey, J.R., "Solid-State Techniques for Improving Solubility," AAPS Advances in Pharmaceutical Sciences Series (D. Crommelin ed., 2012).
Jones, E. et al. "Cardioprotective effects in aged spontaneously hypertensive rats due to chronic stimulation/activation of sGC without hypotension," BMC Pharmacol. 2009, p. 29, 9. [abstract].
Jungmann, N.A. et al., "In vitro-in vivo correlation of the drug-drug interaction potential of antiretroviral HIV treatment regimens on CYP1A1 substrate riociguat," Expert Opinion on Drug Metabolism & Toxicology, 2019, 15(11), 975-984.
Kim at al., "Cirsium japonicumelicits endothelium-dependent relaxation via histamine H1-receptor in rat thoracic aorta," Journal of Ethno-Pharmacology, 2008, 116: 223-227.
Ko et al., "YC-1, a Novel Activator of Platelet Guanylate Cyclase," Blood, Dec. 1994, 84(12): 4226-4233.
Kratochvil, B., "Solid Forms of Pharmaceutical Molecules," Glassy, ch. 8 Amorphous and Nano-Crystalline Materials, J. Simon ed 2011 pp. 129-140.
Lang, D. et al., "Highly Variable Expression of CYP1A1 in Human Liver and Impact on Pharmacokinetics of Riociguat and Granisetron in Humans," Chem. Res. Toxicol. 2019, 32, 1115-1122.
Li et al., "Synthesis and Structure-Activity Relationships of 2-Pyridones: A Novel Series of Potent DNA Gyrase Inhibitors as Antibacterial Agents," J Med. Chem., 1996, 39: 3070-3088.
Markovski et al., "Reactions of Pentakis(2,2,3,3-Tetrafluoropropoxy) Phosphorane with Secondary Amines," Zhurnal Obshchei Khimii 1980, 50(4):826-834.
Masuyama, H. et al. "Pressure-independent effects of pharmacological stimulation of soluble guanylate cyclase on fibrosis in pressure-overloaded rat heart," Hypertens Res. 2009, 597-603, 32.
Masuyama, H. et al. "Soluble guanylate cyclase stimulation on cardiovascular remodeling in angiotensin II-induced hypertensive rats," Hypertension, 2006, 972-978, 48.
Methner, C. et al. "Riociguat Reduces Infarct Size and Post-Infarct Heart Failure in Mouse Hearts: Insights from MRI/PET Imaging," Dec. 2013, 1-8, vol. 8, Issue 12, e83910.
Mirza, S. et al., "Influence of Solvents on the Variety of Crystalline Forms of Erythromycin", AAPS PharmSci 2003; 9 pages.
Mittendorf et al., "Discovery of Riociguat (BAY 63-2521): A Potent, Oral Stimulator of Soluble Guanylate Cyclase for the Treatment of Pulmonary Hypertension," Chem. Med. Chem., 2009, 4: 853-865.
Morissette, S. et al., "High-throughput crystallization: polymorphs, salts, co-crystals and solvates of pharmaceutical solids", Elsevier, Advanced Drug Delivery Reviews 56 (2004); pp. 275-300.
Mulsch et al., "Effect of YC-1, an NO-independent, superoxide-sensitive stimulator of soluble guanylyl cyclase, on smooth muscle responsiveness to nitrovasodilators," Brit. J. Pharm., 1997, 120: 681-689.
Palacios et al. "A New and Efficient Synthesis of Imidazo[1,5-a]pyridine Derivatives by a Tandem Aza-Wittig/Electrocyclic Ring Closure of N-vinylic phosphazenes," Tetrahedron, 1995, 51(12): 3683-3690.
Patani, G.A. et al., "Bioisosterism: A Rational Approach in Drug Design," Chemical Reviews, 1996, pp. 3147-3176, vol. 96, No. 8, American Chemical Society, Washington DC, USA.
Pettibone et al., "A Structurally Novel Stimulator of Guanylate Cyclase with Long-Lasting Hypotensive Activity in the Dog," Euro. J. of Pharmacology, 1985, 116: 307-312.
Pieske, B. "Vericiguat in patients with worsening chronic heart failure and preserved ejection fraction: results of the soluble guanylate cyclase stimulator in heart failure patients with preserved EF (Socrates-Preserved) study," European Heart Journal, 2017, 1119-1127, 38.
Powers-Martin et al., "Immunohistochemical assessment of cyclic guanosine monophosphate (cGMP) and soluble guanylate cyclase (sGC) within the rostral ventrolateral medulla," J. Biomed Sci., 2008, 150: 801-812.
Reichardt et al., "Darstellung von Fluor-und Jodmalondialdehyd," Liebigs Ann. Chem., 1970, 737: 99-107.
Schwoch et al., "189. 2-3-Dihydrospirol [1H-4 and 5-azabenzimidazole-2,1′-cyclohexane](=Spiro [cyclohexane-1,2′(3′H)-1′H-imidazo[4,5-hb]pyridine] and Spiro[cyclohexane-1,2′(3′H)-1′H-imidazo[4,5-c[pyridine]): Reactions with Nucleophiles," Helvetia Chimica Acta, 1994, 77: 2175-2190.
Sharkovska et al., "Nitric oxide-independent stimulation of soluble guanylate cyclase reduces organ damage In experimental low-renin and high-renin models," J. Hypertnesion, 2010, 28(8):1666-1675.
Sharkovska, Y. et al. "Nitric oxide-independent stimulation of soluble guanylate cyclase reduces organ damage in experimental low-renin and high-renin models," Journal of Hypertension 2010, 1666-1675, 28.
Stasch et al., "Soluble Guanylate Cyclase as an Emerging Therapeutic Target In Cardiopulmonary Disease," Circulation, May 2011, 123: 2263-2273.
Stasch, J. et al. "Renal effects of soluble guanylate cyclase stimulators and activators: A review of the preclinical evidence," Current Opinion in Pharmacology 2015, 95-104, 21.
Storey et al. eds., (2011) Solid State Characterization of Pharmaceuticals.
Van Der Mooter, G. "The use of amorphous solid dispersions: A formulation strategy to overcome poor solubility and dissolution rate," Drug Discovery Today: Technologies, vol. 9 No. 2 (2012), e79-e85.
Vippagunta et al., "Crystalline solids," Advanced Drug Delivery Reviews, 2001, 48: 3-26.
Vogt, F. et al., "A study of variable hydration states in topotecan hydrochloride", Elsevier Journal of Pharmaceutical and Biomedical Analysis 40 (2006); pp. 1080-1088.
Wang, Y. et al. "Enhancing cGMP in experimental progressive renal fibrosis: soluble guanylate cyclase stimulation vs. phosphodiesterase inhibition," Am J Physiol Renal Physiol, 2006, F167-F176, 290.
Wang, Y. et al. "Stimulation of soluble guanylate cyclase slows progression in anti-thy1-induced chronic glomerulosclerosis," Kidney Int 2005, 47-61, 68.
Winn et al., "2-(Alkylamino)nicotinic Acid and Analogs. Potent Angiotensin II Antagonists," J. Med. Chem 1993, 36: 2676-2688.
Wu et al., "YC-1 inhibited human platelet aggregation through NO-independent activation of soluble guanylate cyclase," Br J. Pharmacol. Oct. 1995, 116(3): 1973-1978.
Wunder et al., "A cell-based cGMP assay useful for ultra-high-throughput screening and Identification of modulators of the nitric oxide/cGMP pathway," Anal. Biochem., 2005, 339:104-112.
Yamanaka et al., "A New Facile and Efficient Method for the Preparation of (Z)-α-Fluoro-β-(dialkylamino) acrylaldehydes from (Polyfluoro-1-propenyl)trimethylammonium Iodide," Synlett, May 1993, 353-354.
Yamanaka et al., "Reactions of Polyfluoroalkyl o-Nitrobenzenesulfonates with Tertiary Amines," Nippon Kagaku Kaishi 1985, 10: 1988-1994 (in JP + abstract).
Yamanaka et al., "Synthesis and Reactions of (1H, 1H, αH-Perfluoroalkyl)-trimethylammonium Halides," Nippon Kagaku Kaishi, 1988, 7: 1036-1043 (in JP+abstract).
Yu et al., "Vasorelaxant Effect of Isoliquiritigenin, A novel soluble guanylate cyclase activator, in rat aorta," Brit. J. of Pharmacology, 1995, 114: 1587-1594.
Zhao et al., "Effect of aspirin, clopidogrel, and dipyridamole on soluble markers of vascular function in normal volunteers and patients with prior ischaemic stroke," Platelets, 2006,17(2):100-104.

Also Published As

Publication number Publication date
NZ603800A (en) 2014-10-31
TN2012000549A1 (en) 2014-04-01
JP5896991B2 (en) 2016-03-30
BR112012030010A2 (en) 2020-09-01
IL223050A (en) 2017-01-31
US20160129004A1 (en) 2016-05-12
CA2800697C (en) 2018-08-07
UA109660C2 (en) 2015-09-25
JP2013530150A (en) 2013-07-25
FR21C1038I2 (en) 2022-09-02
US20150080414A1 (en) 2015-03-19
AU2011257335A1 (en) 2012-12-20
LTPA2021518I1 (en) 2021-09-10
LTC2576547I2 (en) 2022-10-25
EP2576547A1 (en) 2013-04-10
IL249669B (en) 2020-05-31
TW201210592A (en) 2012-03-16
SI2576547T1 (en) 2016-05-31
JO3199B1 (en) 2018-03-08
HUS2100032I1 (en) 2021-09-28
US20200397785A1 (en) 2020-12-24
FR21C1038I1 (en) 2021-10-15
ME02393B (en) 2016-09-20
PL2576547T3 (en) 2016-07-29
US20130237551A1 (en) 2013-09-12
US20170273977A1 (en) 2017-09-28
GT201200313A (en) 2014-01-31
CU24086B1 (en) 2015-03-30
CY1117372T1 (en) 2017-04-26
AR081628A1 (en) 2012-10-10
EP2576547B1 (en) 2016-01-27
EA023631B9 (en) 2017-04-28
US8420656B2 (en) 2013-04-16
CO6640226A2 (en) 2013-03-22
DE102010021637A1 (en) 2011-12-01
BR122020023470B1 (en) 2022-07-12
MY164356A (en) 2017-12-15
US10736896B2 (en) 2020-08-11
CR20120596A (en) 2013-04-22
CU20120161A7 (en) 2013-02-26
HUE026912T2 (en) 2016-07-28
NL301146I1 (en) 2021-12-08
MX2012013324A (en) 2012-12-05
KR20130116000A (en) 2013-10-22
HRP20160370T1 (en) 2016-05-06
DOP2012000299A (en) 2013-03-31
NL301146I2 (en) 2022-07-06
US20180263981A1 (en) 2018-09-20
US9993476B2 (en) 2018-06-12
RS54704B1 (en) 2016-08-31
KR101862420B1 (en) 2018-07-04
US20120022084A1 (en) 2012-01-26
CN102939289A (en) 2013-02-20
US20220409618A1 (en) 2022-12-29
WO2011147809A1 (en) 2011-12-01
CN102939289B (en) 2016-01-20
MA34248B1 (en) 2013-05-02
IL249669A0 (en) 2017-02-28
SG185460A1 (en) 2012-12-28
PE20130402A1 (en) 2013-04-13
HK1182112A1 (en) 2013-11-22
UY33396A (en) 2011-12-30
AU2011257335B2 (en) 2015-12-03
DK2576547T3 (en) 2016-04-25
IL223050A0 (en) 2013-02-03
ECSP12012310A (en) 2012-12-28
CL2012003226A1 (en) 2013-07-26
US8921377B2 (en) 2014-12-30
ES2567795T3 (en) 2016-04-26
CA2800697A1 (en) 2011-12-01
EA023631B1 (en) 2016-06-30
TWI516263B (en) 2016-01-11
NO2021032I1 (en) 2021-09-06
US9266885B2 (en) 2016-02-23
EA201291336A1 (en) 2013-04-30

Similar Documents

Publication Publication Date Title
US11439642B2 (en) Substituted 5-fluoro-1H-pyrazolopyridines and their use
US9309239B2 (en) Substituted 6-fluoro-1H-pyrazolo[4,3-b]pyridines and use thereof
US9266871B2 (en) Trifluoromethyl-substituted fused pyrimidines and their use
US8765769B2 (en) Ring-fused 4-aminopyrimidines and use thereof as stimulators of soluable guanylate cyclases
US9090610B2 (en) Fluoroalkyl-substituted pyrazolopyridines and use thereof
US9498480B2 (en) Substituted azabicycles and use thereof
US9133191B2 (en) Substituted triazine derivatives and use thereof as stimulators of soluble guanylate cyclase
US20140228366A1 (en) Heteroaryl-substituted pyrazolopyridines and use thereof as soluble guanylate cyclase stimulators
US9605008B2 (en) Benzyl-1H-pyrazolo[3,4-b]pyridines and use thereof
US9150580B2 (en) Substituted imidazopyridines and the use thereof
US20140128425A1 (en) Carboxy-substituted imidazo[1,2-a]pyridinecarboxamides and their use
CA2902987A1 (en) Benzyl-substituted pyrazolopyridines and use thereof

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: BAYER PHARMA AKTIENGESELLSCHAFT, GERMANY

Free format text: CHANGE OF NAME;ASSIGNOR:BAYER SCHERING PHARMA AKTIENGESELLSCHAFT;REEL/FRAME:055222/0814

Effective date: 20110701

Owner name: ADVERIO PHARMA GMBH, GERMANY

Free format text: NUNC PRO TUNC ASSIGNMENT;ASSIGNOR:BAYER INTELLECTUAL PROPERTY GMBH;REEL/FRAME:055152/0361

Effective date: 20141223

Owner name: BAYER INTELLECTUAL PROPERTY GMBH, GERMANY

Free format text: NUNC PRO TUNC ASSIGNMENT;ASSIGNOR:BAYER PHARMA AKTIENGESELLSCHAFT;REEL/FRAME:055152/0031

Effective date: 20120401

Owner name: BAYER SCHERING PHARMA AKTIENGESELLSCHAFT, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FOLLMANN, MARKUS;STASCH, JOHANNES-PETER;REDLICH, GORDEN;AND OTHERS;SIGNING DATES FROM 20110606 TO 20110620;REEL/FRAME:055151/0491

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE